Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Bacteriol ; 201(11)2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30782639

RESUMEN

Removal of one acyl chain from bacterial lipid A by deacylase activity is a mechanism used by many pathogenic bacteria to evade the host's Toll-like receptor 4 (TLR4)-mediated innate immune response. In Porphyromonas gingivalis, a periodontal pathogen, lipid A deacylase activity converts a majority of the initially synthesized penta-acylated lipid A, a TLR4 agonist, to tetra-acylated structures, which effectively evade TLR4 sensing by being either inert or antagonistic at TLR4. In this paper, we report successful identification of the gene that encodes the P. gingivalis lipid A deacylase enzyme. This gene, PGN_1123 in P. gingivalis 33277, is highly conserved within P. gingivalis, and putative orthologs are phylogenetically restricted to the Bacteroidetes phylum. Lipid A of ΔPGN_1123 mutants is penta-acylated and devoid of tetra-acylated structures, and the mutant strain provokes a strong TLR4-mediated proinflammatory response, in contrast to the negligible response elicited by wild-type P. gingivalis Heterologous expression of PGN_1123 in Bacteroides thetaiotaomicron promoted lipid A deacylation, confirming that PGN_1123 encodes the lipid A deacylase enzyme.IMPORTANCE Periodontitis, commonly referred to as gum disease, is a chronic inflammatory condition that affects a large proportion of the population. Porphyromonas gingivalis is a bacterium closely associated with periodontitis, although how and if it is a cause for the disease are not known. It has a formidable capacity to dampen the host's innate immune response, enabling its persistence in diseased sites and triggering microbial dysbiosis in animal models of infection. P. gingivalis is particularly adept at evading the host's TLR4-mediated innate immune response by modifying the structure of lipid A, the TLR4 ligand. In this paper, we report identification of the gene encoding lipid A deacylase, a key enzyme that modifies lipid A to TLR4-evasive structures.


Asunto(s)
Proteínas Bacterianas/genética , Hidrolasas de Éster Carboxílico/genética , Regulación Bacteriana de la Expresión Génica , Evasión Inmune/genética , Lípido A/química , Porphyromonas gingivalis/genética , Receptor Toll-Like 4/genética , Carga Bacteriana , Proteínas Bacterianas/metabolismo , Bacteroides thetaiotaomicron/genética , Bacteroides thetaiotaomicron/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Línea Celular , Secuencia Conservada , Células HEK293 , Humanos , Lípido A/inmunología , Monocitos/inmunología , Monocitos/microbiología , Porphyromonas gingivalis/metabolismo , Receptor Toll-Like 4/inmunología
2.
Infect Immun ; 87(12)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31570556

RESUMEN

The Porphyromonas gingivalis strain ATCC 33277 (33277) and 381 genomes are nearly identical. However, strain 33277 displays a significantly diminished capacity to stimulate host cell Toll-like receptor 2 (TLR2)-dependent signaling and interleukin-1ß (IL-1ß) production relative to 381, suggesting that there are strain-specific differences in one or more bacterial immune-modulatory factors. Genomic sequencing identified a single nucleotide polymorphism in the 33277 fimB allele (A→T), creating a premature stop codon in the 33277 fimB open reading frame relative to the 381 fimB allele. Gene exchange experiments established that the 33277 fimB allele reduces the immune-stimulatory capacity of this strain. Transcriptome comparisons revealed that multiple genes related to carboxy-terminal domain (CTD) family proteins, including the gingipains, were upregulated in 33277 relative to 381. A gingipain substrate degradation assay demonstrated that cell surface gingipain activity is higher in 33277, and an isogenic mutant strain deficient for the gingipains exhibited an increased ability to induce TLR2 signaling and IL-1ß production. Furthermore, 33277 and 381 mutant strains lacking CTD cell surface proteins were more immune-stimulatory than the parental wild-type strains, consistent with an immune-suppressive role for the gingipains. Our data show that the combination of an intact fimB allele and limited cell surface gingipain activity in P. gingivalis 381 renders this strain more immune-stimulatory. Conversely, a defective fimB allele and high-level cell surface gingipain activity reduce the capacity of P. gingivalis 33277 to stimulate host cell innate immune responses. In summary, genomic and transcriptomic comparisons identified key virulence characteristics that confer divergent host cell innate immune responses to these highly related P. gingivalis strains.


Asunto(s)
Proteínas Fimbrias/genética , Proteínas Fimbrias/inmunología , Cisteína-Endopeptidasas Gingipaínas/metabolismo , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/inmunología , Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/microbiología , Línea Celular Tumoral , Células HEK293 , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Interleucina-1beta/metabolismo , Polimorfismo de Nucleótido Simple/genética , Transducción de Señal/inmunología , Células THP-1 , Receptor Toll-Like 2/metabolismo
3.
Infect Immun ; 81(4): 1277-86, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23381996

RESUMEN

Infection by the chronic periodontitis-associated pathogen Porphyromonas gingivalis activates a Toll-like receptor 2 (TLR2) response that triggers inflammation in the host but also promotes bacterial persistence. Our aim was to define ligands on the surfaces of intact P. gingivalis cells that determine its ability to activate TLR2. Molecules previously reported as TLR2 agonists include lipopolysaccharide (LPS), fimbriae, the lipoprotein PG1828, and phosphoceramides. We demonstrate that these molecules do not comprise the major factors responsible for stimulating TLR2 by whole bacterial cells. First, P. gingivalis mutants devoid of the reported protein agonists, PG1828 and fimbriae, activate TLR2 as strongly as the wild type. Second, two-phase extraction of whole bacteria resulted in a preponderance of TLR2 agonist activity partitioning to the hydrophilic phase, demonstrating that phosphoceramides are not a major TLR2 ligand. Third, analysis of LPS revealed that TLR2 activation is independent of lipid A structural variants. Instead, activation of TLR2 and TLR2/TLR1 by LPS is in large part due to copurifying molecules that are sensitive to the action of the enzyme lipoprotein lipase. Strikingly, intact P. gingivalis bacterial cells treated with lipoprotein lipase were attenuated in their ability to activate TLR2. We propose that a novel class of molecules comprised by lipoproteins constitutes the major determinants that confer to P. gingivalis the ability to stimulate TLR2 signaling.


Asunto(s)
Lipoproteína Lipasa/metabolismo , Porphyromonas gingivalis/enzimología , Porphyromonas gingivalis/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Línea Celular , Humanos , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Lipoproteínas/metabolismo , Porphyromonas gingivalis/patogenicidad , Transducción de Señal , Factores de Virulencia/metabolismo
4.
Infect Immun ; 79(1): 203-10, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20974832

RESUMEN

The human symbiont Bacteroides thetaiotaomicron promotes intestinal function and health, whereas the phylogenetically related pathogen Porphyromonas gingivalis is associated with the chronic oral inflammatory disease periodontitis. Although both B. thetaiotaomicron and P. gingivalis synthesize lipopolysaccharides (LPS) consisting of penta-acylated, monophosphorylated lipid A in addition to immunologically silent, nonphosphorylated lipid A, they elicit strikingly distinct Toll-like receptor 4 (TLR4) responses. We show that the phosphate position of penta-acylated, monophosphorylated lipid A is a key feature for determining the differential TLR4 responses elicited by these evolutionarily related bacteria. B. thetaiotaomicron produces TLR4-stimulatory lipid A bearing a 1-phosphate, in contrast to P. gingivalis, which produces TLR4-evasive lipid A bearing a 4'-phosphate. Confirming these observations, recombinant Escherichia coli LPS containing penta-acylated, 1-phosphorylated lipid A is more TLR4 stimulatory than LPS containing 4'-phosphorylated lipid A. The specific capacity of a Gram-negative bacterium to alert or evade the host innate immune defense system through TLR4-dependent signaling is currently recognized as a critical aspect defining the relationship between the host and the bacterium. We propose that the distinct lipid A phosphate positions observed for the B. thetaiotaomicron and P. gingivalis LPS contributes to the manifestation of these bacteria as commensal or pathogen within the human host.


Asunto(s)
Bacteroides/genética , Escherichia coli/genética , Lípido A/química , Porphyromonas gingivalis/genética , Receptor Toll-Like 4/metabolismo , Bacteroides/metabolismo , Conformación de Carbohidratos , Escherichia coli/metabolismo , Regulación de la Expresión Génica/fisiología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Lípido A/metabolismo , Filogenia , Porphyromonas gingivalis/metabolismo , Simbiosis , Receptor Toll-Like 4/genética
5.
Front Oral Health ; 2: 689475, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35048035

RESUMEN

Aim: Clinically healthy gingival tissue is maintained through controlled regulation of host defense mechanisms against plaque biofilm overgrowth. One key component is the transit of neutrophils from the vasculature into gingival tissue where the expression of different neutrophil chemokines are tightly regulated. This cross-sectional study examines the inter-individual variability in chemokine profiles within gingival crevicular fluid (GCF) in relation to the subgingival bacterial community in a state of gingival health. Methods: Gingival crevicular fluid and subgingival plaque samples were collected from mesiobuccal surfaces of all six Ramfjord teeth of 20 systemically healthy individuals (14.55 ± 1.67 years). A multiplex immunoassay was carried out to quantify the expression of 40 different chemokines in the healthy gingival tissue. Neutrophils were assessed indirectly by myeloperoxidase (MPO) in GCF using traditional ELISA. Characterization of healthy subgingival plaque was conducted with the Illumina Miseq targeting the 16S rRNA gene. Results: In health, there are distinct variations within individual gingival crevicular fluid chemokine expression profiles, as well as in the concentration of neutrophils, that divided the participants into high or low chemokine expressing groups. Specifically, key differences were identified within MIF (2683.54 ± 985.82 pg per 30-s sample), IL-8/CXCL8 (170.98 ± 176.96 pg per 30-s sample), Gro-α/CXCL1 (160.42 ± 94.21 pg per 30-s sample), ENA-78/CXCL5 (137.76 ± 76.02 pg per 30-s sample), IL-1ß (51.39 ± 37.23 pg per 30-s sample), TNF-α (1.76 ± 1.79 pg per 30-s sample), and IFN-γ (0.92 ± 0.54 pg per 30-s sample). Of these identified chemokines, the highest correlation was associated between IL-8/CXCL8 and neutrophils (r = 0.54, p = 0.014). Furthermore, species characterization of healthy subgingival plaque revealed significant inter-individual variability that identified two unique groups unrelated to the previously identified chemokine groups. Conclusion: The lack of concordance between the microbial composition and chemokine profile during health may be a reflection of the unique microbial composition of each individual coupled with variations within their host response, emphasizing the vast complexity of the defense mechanisms in place to maintain gingival health.

7.
Clin Implant Dent Relat Res ; 17(6): 1029-35, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24909337

RESUMEN

BACKGROUND: There is little consensus on the most appropriate cement to use when restoring a cement-retained, implant-supported restoration. One consideration should be the interaction of pathogenic oral bacteria with restorative cements. PURPOSE: To determine how oral bacteria associated with peri-implant disease grow in the presence of implant cements. MATERIALS AND METHODS: Five test cements with varying composition (zinc oxide-eugenol [TBO], eugenol-free zinc oxide [TBNE], zinc orthophosphate [FL], and two resin cements [PIC and ML]) were used to fabricate specimen disks. The disks were submerged in bacterial suspensions of either Aggregatibacter actinomycetemcomitans, Fusobacterium nucleatum, or Porphyromonas gingivalis. Planktonic bacterial growth within the test media was measured by determining the optical density of the cultures (OD600 ). Positive controls (media and bacteria without cement disks) and negative controls (media alone) were similarly evaluated. The mean and standard deviations (SD) were calculated for planktonic growth from three separate experiments. ANOVA statistical analysis with post hoc Tukey tests was performed where differences existed (p < .05). Selected cement disks (TBO and ML) were further examined for bacterial biofilm growth. Surface bacteria were removed and grown on agar media, and colony-forming units (CFUs) were quantified. RESULTS: Planktonic growth for both A. actinomycetemcomitans and P. gingivalis was significantly inhibited (p < .05) when grown in the presence of cement disks consisting of TBNE, PIC, FL, and TBO. In contrast, neither of these bacteria displayed growth inhibition in the presence of ML cement disks. F. nucleatum growth was also significantly inhibited by PIC, FL. and TBO (p < .05), but not by ML and TBNE cement disks. CFU counts for the biofilm study for TBO gave minimal and, in some instances, no bacterial adherence and growth, in contrast to ML, which supported substantially greater bacterial biofilm growth. CONCLUSION: Cements display differing abilities to inhibit both planktonic and biofilm bacterial growth. Cements with the ability to reduce planktonic or biofilm growth of the test bacteria may be advantageous in reducing peri-implant disease. Understanding the microbial growth-inhibiting characteristics of different cement types should be considered important in the selection criteria.


Asunto(s)
Bacterias/crecimiento & desarrollo , Biopelículas/crecimiento & desarrollo , Cementos Dentales/farmacología , Prótesis Dental de Soporte Implantado , Periimplantitis/microbiología , Eugenol , Técnicas In Vitro , Cementos de Resina/farmacología , Óxido de Zinc , Cemento de Óxido de Zinc-Eugenol/farmacología , Cemento de Fosfato de Zinc/farmacología
8.
Quintessence Int ; 43(8): 703-7, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23034423

RESUMEN

OBJECTIVE: To microbiologically evaluate the efficacy of cotton and polytetrafluoroethylene (PTFE) tape used as spacer materials. METHOD AND MATERIALS: Twenty-six extracted human molars were restored using either cotton or PTFE tape as spacers under a standardized provisional restorative material (Cavit). The teeth were incubated for 7 days in a culture of Streptococcus gordonii or in liquid media alone. The spacers were removed and tested for bacterial contamination. The access cavities were also evaluated for bacterial contamination. RESULTS: Nine of 10 teeth with cotton spacers and one of 10 teeth with PTFE spacers were positive for S gordonii growth. The nine teeth in the cotton group also showed contamination of the access cavities. CONCLUSION: Even under optimal conditions, cotton spacers may cause leakage into the access cavities. Cotton fibers may serve as a route for bacterial contamination of the access cavities and root canal space. In contrast, PTFE tape did not provide an avenue for bacterial contamination.


Asunto(s)
Fibra de Algodón , Filtración Dental/microbiología , Politetrafluoroetileno , Tratamiento del Conducto Radicular , Humanos , Streptococcus gordonii/crecimiento & desarrollo
9.
Int J Oral Sci ; 1(3): 126-35, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20657724

RESUMEN

AIM: To elucidate the genetic basis for the pronounced resistance that the oral pathogen, Porphyromonas gingivalis (P. gingivalis), exhibits towards the cationic antimicrobial peptide, polymyxin B. METHODOLOGY: A genetic screen of P. gingivalis clones generated by a Tn4400'-based random insertion mutagenesis strategy was performed to identify bacteria harboring novel genetic mutations that render P. gingivalis susceptible to killing by the cationic antimicrobial peptide, polymyxin B (PMB, 50 microg x mL(-1)). RESULTS: P. gingivalis (ATCC 33277) is unusually resistant to the cationic antimicrobial peptide, PMB at relatively high concentrations (200 microg x mL(-1)). Approximately 2,700 independent Tn4400'-derived mutants of P. gingivalis were examined for increased sensitivity to PMB killing at a relatively low dose (50 microg x mL(-1)). A single PMB-sensitive mutant was obtained in this phenotypic screen. We determined that the Tn4400' transposon was integrated into the gene encoding the lipid A 4'-phosphatase, PGN_0524, demonstrating that this insertion event was responsible for its increased susceptibility of this clone to PMB-dependent killing. The resulting mutant strain, designated 0524-Tn4400', was highly sensitive to PMB killing relative to wild-type P. gingivalis, and exhibited the same sensitivity as the previously characterized strain, 0524KO, which bears a genetically engineered deletion in the PGN_0524 locus. Positive ion mass spectrometric structural (MALDI-TOF MS) analyses revealed that lipid A isolates from 0524-Tn4400' and 0524KO strains displayed strikingly similar MALDI-TOF MS spectra that were substantially different from the wildtype P. gingivalis lipid A spectrum. Finally, intact 0524-Tn4400' and 0524KO mutant bacteria, as well as their corresponding LPS isolates, were significantly more potent in stimulating Toll-like receptor 4 (TLR4)-dependent E-selectin expression in human endothelial cells relative to intact wild-type P. gingivalis or its corresponding LPS isolate. CONCLUSION: The combined molecular evidence provided in this report suggests that PGN_0524, a lipid A 4'-phosphatase, is the sole genetic element conferring the ability of the periodontopathogen, P. gingivalis, to evade the killing activity of cationic antimicrobial peptides, such as PMB. These data strongly implicate PGN_0524 as a critical virulence factor for the ability of P. gingivalis to evade front-line host innate defenses that are dependent upon cationic antimicrobial peptide activity and TLR 4 sensing.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Monoéster Fosfórico Hidrolasas/fisiología , Polimixina B/farmacología , Porphyromonas gingivalis/enzimología , Mapeo Cromosómico , Elementos Transponibles de ADN/genética , Selectina E/análisis , Selectina E/inmunología , Células Endoteliales/inmunología , Células Endoteliales/microbiología , Eliminación de Gen , Humanos , Lípido A/análisis , Lípido A/inmunología , Lipopolisacáridos/análisis , Lipopolisacáridos/inmunología , Mutagénesis Insercional/genética , Sistemas de Lectura Abierta/genética , Monoéster Fosfórico Hidrolasas/genética , Porphyromonas gingivalis/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Receptor Toll-Like 4/análisis , Receptor Toll-Like 4/inmunología , Factores de Virulencia/fisiología
10.
Microbiology (Reading) ; 154(Pt 6): 1605-1615, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18524915

RESUMEN

Mucoid strains of Pseudomonas aeruginosa that overproduce alginate are associated with chronic pulmonary disease (e.g. cystic fibrosis). Mutants defective in one of several periplasmic proteins (AlgKGX) for alginate secretion release alginate fragments due to the activity of an alginate lyase (AlgL) in the periplasm, which cleaves the newly formed polymers. However, mutants defective in Alg8 or Alg44 did not secrete polymer or alginate fragments, suggesting that both these membrane proteins have a role in the polymerization reaction. A model for the membrane topology of Alg8, a glycosyltransferase (GT), was constructed using PhoA fusions. This provided evidence for a large cytoplasmic loop containing the active domains predicted for beta-GTs such as Alg8 and five transmembrane (TM) domains, one of which resembles a cleavable signal peptide. The C-terminal TM domain of Alg8 was critical for the polymerization reaction in vivo. Alanine substitution mutagenesis showed that all of the predicted active site residues in the widely spaced D, DxD, D, LxxRW motif were required for polymerization activity in vivo, and two of these substitutions also affected Alg8 protein stability. A membrane topology model for Alg44 was also constructed using PhoA fusions, and this showed a central TM domain and predicted an N-terminal TM domain that may be a membrane anchor. An N-terminal PilZ domain in Alg44 for c-di-GMP [bis-(3',5')-cyclic dimeric GMP] binding, which is required for alginate synthesis, was localized to the cytoplasmic loop. The long periplasmic C terminus of Alg44 contains a region similar to membrane fusion proteins (MFPs) of multi-drug efflux systems, which predicts the possibility of its interaction with another protein in this compartment. A Western blot analysis of the outer-membrane porin AlgE showed reduced AlgE levels in the alg44 mutant, whereas expression of Alg44 in trans restored AlgE within the cell. C-terminal truncations of Alg44 as small as 24 amino acids blocked alginate polymerization in vivo, indicating a critical role for the MFP domain. These studies suggest that Alg44 may act as a co-polymerase in concert with Alg8, the major GT, and that both inner-membrane proteins are required in vivo for the polymerization reaction leading to alginate production.


Asunto(s)
Alginatos/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Pseudomonas aeruginosa/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Proteínas Bacterianas/genética , Membrana Celular/metabolismo , Escherichia coli/genética , Ácido Glucurónico/metabolismo , Ácidos Hexurónicos/metabolismo , Proteínas de la Membrana/genética , Polímeros/metabolismo , Porinas/metabolismo , Pseudomonas aeruginosa/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido
11.
J Bacteriol ; 185(12): 3515-23, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12775688

RESUMEN

Bacterial alginates are produced as 1-4-linked beta-D-mannuronan, followed by epimerization of some of the mannuronic acid residues to alpha-L-guluronic acid. Here we report the isolation of four different epimerization-defective point mutants of the periplasmic Pseudomonas fluorescens mannuronan C-5-epimerase AlgG. All mutations affected amino acids conserved among AlgG-epimerases and were clustered in a part of the enzyme also sharing some sequence similarity to a group of secreted epimerases previously reported in Azotobacter vinelandii. An algG-deletion mutant was constructed and found to produce predominantly a dimer containing a 4-deoxy-L-erythro-hex-4-enepyranosyluronate residue at the nonreducing end and a mannuronic acid residue at the reducing end. The production of this dimer is the result of the activity of an alginate lyase, AlgL, whose in vivo activity is much more limited in the presence of AlgG. A strain expressing both an epimerase-defective (point mutation) and a wild-type epimerase was constructed and shown to produce two types of alginate molecules: one class being pure mannuronan and the other having the wild-type content of guluronic acid residues. This formation of two distinct classes of polymers in a genetically pure cell line can be explained by assuming that AlgG is part of a periplasmic protein complex.


Asunto(s)
Alginatos/metabolismo , Carbohidrato Epimerasas/metabolismo , Polímeros/metabolismo , Pseudomonas fluorescens/metabolismo , Secuencia de Aminoácidos , Carbohidrato Epimerasas/genética , Eliminación de Gen , Ácido Glucurónico , Ácidos Hexurónicos , Datos de Secuencia Molecular , Mutación Puntual , Pseudomonas fluorescens/química , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA