Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Bacteriol ; 204(12): e0031322, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36448787

RESUMEN

The periodontal pathogen Tannerella forsythia expresses a ß-glucanase (TfGlcA) whose expression is induced in response to Fusobacterium nucleatum, a bridge bacterium of the oral cavity. TfGlcA cleaves ß-glucans to release glucose, which can serve as a carbon source for F. nucleatum and other cohabiting organisms. A two-gene cluster encoding a putative extracytoplasmic function (ECF) sigma factor and a FecR-like anti-sigma factor has been recognized upstream of a TfGlcA operon. We characterized and analyzed the role of these putative ECF sigma and anti-sigma factors in the regulation of TfGlcA expression. For this purpose, deletion mutants were constructed and analyzed for ß-glucanase expression. In addition, an Escherichia coli-produced ECF sigma factor recombinant protein was evaluated for transcriptional and DNA binding activities. The results showed that the recombinant protein promoted transcription by the RNA polymerase core enzyme from the glcA promoter. Furthermore, in comparison to those in the parental strain, the ß-glucanase expression levels were significantly reduced in the ECF sigma-factor deletion mutant and increased significantly in the FecR anti-sigma factor deletion mutant. The levels did not change in the mutants following coincubation with the F. nucleatum whole cells or cell extracts. Finally, the levels of ß-glucanase produced by T. forsythia strains paralleled F. nucleatum biomass in cobiofilms. In conclusion, we identified a ß-glucanase operon regulatory system in T. forsythia comprising an ECF sigma factor (TfSigG) and a cognate FecR-like anti-sigma factor responsive to F. nucleatum and potentially other stimuli. IMPORTANCE Previous studies have shown that F. nucleatum forms robust biofilms with T. forsythia utilizing glucose from the hydrolysis of ß-glucans by T. forsythia ß-glucanase, induced by F. nucleatum. In this study, we showed that a regulatory system comprising of an ECF sigma factor, TfSigG, and a FecR-like anti-sigma factor, TfFecR, is responsible for the ß-glucanase induction in response to F. nucleatum, suggesting that this system plays roles in the mutualistic interactions of T. forsythia and F. nucleatum. The findings suggest the development and potential utility of small-molecule inhibitors targeting the ß-glucanase activity or the TfSigG/TfFecR system as therapeutic drugs against dental plaque formation and periodontitis.


Asunto(s)
Fusobacterium nucleatum , Glucosidasas , Tannerella forsythia , Biopelículas , Fusobacterium nucleatum/genética , Fusobacterium nucleatum/metabolismo , Factor sigma/genética , Factor sigma/metabolismo , Glucosidasas/genética
2.
Microbiology (Reading) ; 165(11): 1181-1197, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31517596

RESUMEN

Key to onset and progression of periodontitis is a complex relationship between oral bacteria and the host. The organisms most associated with severe periodontitis are the periodontal pathogens of the red complex: Tannerella forsythia, Treponema denticola and Porphyromonas gingivalis. These organisms express sialidases, which cleave sialic acid from host glycoproteins, and contribute to disease through various mechanisms. Here, we expressed and purified recombinant P. gingivalis sialidase SiaPG (PG_0352) and characterized its activity on a number of substrates, including host sialoglycoproteins and highlighting the inability to cleave diacetylated sialic acids - a phenomenon overcome by the NanS sialate-esterase from T. forsythia. Indeed SiaPG required NanS to maximize sialic acid harvesting from heavily O-acetylated substrates such as bovine salivary mucin, hinting at the possibility of interspecies cooperation in sialic acid release from host sources by these members of the oral microbiota. Activity of SiaPG and P. gingivalis was inhibited using the commercially available chemotherapeutic zanamivir, indicating its potential as a virulence inhibitor, which also inhibited sialic acid release from mucin, and was capable of inhibiting biofilm formation of P. gingivalis on oral glycoprotein sources. Zanamivir also inhibited attachment and invasion of oral epithelial cells by P. gingivalis and other periodontal pathogens, both in monospecies but also in multispecies infection experiments, indicating potential to suppress host-pathogen interactions of a mixed microbial community. This study broadens our understanding of the multifarious roles of bacterial sialidases in virulence, and indicates that their inhibition with chemotherapeutics could be a promising strategy for periodontitis therapy.


Asunto(s)
Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Neuraminidasa/metabolismo , Porphyromonas gingivalis/enzimología , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Línea Celular , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Interacciones Microbianas , Mucinas/metabolismo , Mutación , Neuraminidasa/genética , Polisacáridos/metabolismo , Porphyromonas gingivalis/efectos de los fármacos , Porphyromonas gingivalis/crecimiento & desarrollo , Porphyromonas gingivalis/patogenicidad , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sialoglicoproteínas/metabolismo , Tannerella forsythia/enzimología , Factores de Virulencia/genética , Zanamivir/farmacología
3.
Infect Immun ; 86(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29229734

RESUMEN

Oral streptococci are generally considered commensal organisms; however, they are becoming recognized as important associate pathogens during the development of periodontal disease as well as being associated with several systemic diseases, including as a causative agent of infective endocarditis. An important virulence determinant of these bacteria is an ability to evade destruction by phagocytic cells, yet how this subversion occurs is mostly unknown. Using Streptococcus gordonii as a model commensal oral streptococcus that is also associated with disease, we find that resistance to reactive oxygen species (ROS) with an active ability to damage phagosomes allows the bacterium to avoid destruction within macrophages. This ability to survive relies not only on the ROS resistance capabilities of the bacterium but also on ROS production by macrophages, with both being required for maximal survival of internalized bacteria. Importantly, we also show that this dependence on ROS production by macrophages for resistance has functional significance: S. gordonii intracellular survival increases when macrophages are polarized toward an activated (M1) profile, which is known to result in prolonged phagosomal ROS production compared to that of alternatively (M2) polarized macrophages. We additionally find evidence of the bacterium being capable of both delaying the maturation of and damaging phagosomes. Taken together, these results provide essential insights regarding the mechanisms through which normally commensal oral bacteria can contribute to both local and systemic inflammatory disease.


Asunto(s)
Polaridad Celular , Macrófagos/microbiología , Fagosomas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus gordonii/crecimiento & desarrollo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Humanos , Macrófagos/citología , Macrófagos/inmunología , Ratones , Fagosomas/microbiología , Células RAW 264.7 , Especies Reactivas de Oxígeno/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus gordonii/genética , Streptococcus gordonii/inmunología
4.
Appl Environ Microbiol ; 84(1)2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29079615

RESUMEN

Tannerella forsythia and Fusobacterium nucleatum are dental plaque bacteria implicated in the development of periodontitis. These two species have been shown to form synergistic biofilms and have been found to be closely associated in dental plaque biofilms. A number of genetic loci for TonB-dependent membrane receptors (TDR) for glycan acquisition, with many existing in association with genes coding for enzymes involved in the breakdown of complex glycans, have been identified in T. forsythia In this study, we focused on a locus, BFO_0186-BFO_0188, that codes for a predicted TDR-SusD transporter along with a putative ß-glucan hydrolyzing enzyme (BFO_0186). This operon is located immediately downstream of a 2-gene operon that codes for a putative stress-responsive extracytoplasmic function (ECF) sigma factor and an anti-sigma factor. Here, we show that BFO_0186 expresses a ß-glucanase that cleaves glucans with ß-1,6 and ß-1,3 linkages. Furthermore, the BFO_0186-BFO_0188 locus is upregulated, with an induction of ß-glucanase activity, in cobiofilms of T. forsythia and F. nucleatum The ß-glucanase activity in mixed biofilms in turn leads to an enhanced hydrolysis of ß-glucans and release of glucose monomers and oligomers as nutrients for F. nucleatum In summary, our study highlights the role of T. forsythia ß-glucanase expressed by the asaccharolytic oral bacterium T. forsythia in the development of T. forsythia-F. nucleatum mixed species biofilms, and suggest that dietary ß-glucans might contribute in plaque development and periodontal disease pathogenesis.IMPORTANCE The development of dental plaque biofilm is a complex process in which metabolic, chemical and physical interactions between bacteria take a central role. Previous studies have shown that the dental pathogens T. forsythia and F. nucleatum form synergistic biofilms and are closely associated in human dental plaque. In this study, we show that ß-glucanase from the periodontal pathogen T. forsythia plays a role in the formation of T. forsythia-F. nucleatum cobiofilms by hydrolyzing ß-glucans to glucose as a nutrient. We also unveiled that the expression of T. forsythia ß-glucanase is induced in response to F. nucleatum sensing. This study highlights the involvement of ß-glucanase activity in the development of T. forsythia-F. nucleatum biofilms and suggests that intake of dietary ß-glucans might be a contributing risk factor in plaque development and periodontal disease pathogenesis.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Placa Dental/microbiología , Fusobacterium nucleatum/fisiología , Tannerella forsythia/enzimología , Fusobacterium nucleatum/crecimiento & desarrollo , Humanos
5.
Microb Pathog ; 94: 12-20, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26318875

RESUMEN

Tannerella forsythia is a periodontal pathogen implicated in periodontitis. This gram-negative pathogen depends on exogenous peptidoglycan amino sugar N-acetylmuramic acid (NAM) for growth. In the biofilm state the bacterium can utilize sialic acid (Neu5Ac) instead of NAM to sustain its growth. Thus, the sialic acid utilization system of the bacterium plays a critical role in the growth and survival of the organism in the absence of NAM. We sought the function of a T. forsythia gene annotated as nanT coding for an inner-membrane sugar transporter located on a sialic acid utilization genetic cluster. To determine the function of this putative sialic acid transporter, an isogenic nanT-deletion mutant generated by allelic replacement strategy was evaluated for biofilm formation on NAM or Neu5Ac, and survival on KB epithelial cells. Moreover, since T. forsythia forms synergistic biofilms with Fusobacterium nucleatum, co-biofilm formation activity in mixed culture and sialic acid uptake in culture were also assessed. The data showed that the nanT-inactivated mutant of T. forsythia was attenuated in its ability to uptake sialic acid. The mutant formed weaker biofilms compared to the wild-type strain in the presence of sialic acid and as co-biofilms with F. nucleatum. Moreover, compared to the wild-type T. forsythia nanT-inactivated mutant showed reduced survival when incubated on KB epithelial cells. Taken together, the data presented here demonstrate that NanT-mediated sialic transportation is essential for sialic acid utilization during biofilm growth and survival of the organism on epithelial cells and implies sialic acid might be key for its survival both in subgingival biofilms and during infection of human epithelial cells in vivo.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Células Epiteliales/microbiología , Transportadores de Anión Orgánico/metabolismo , Simportadores/metabolismo , Tannerella forsythia/metabolismo , Adhesión Bacteriana , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , ADN Bacteriano/genética , Fusobacterium nucleatum/crecimiento & desarrollo , Fusobacterium nucleatum/metabolismo , Genes Bacterianos , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/metabolismo , Humanos , Células KB , Ácidos Murámicos/metabolismo , Neuraminidasa/metabolismo , Transportadores de Anión Orgánico/biosíntesis , Transportadores de Anión Orgánico/genética , Eliminación de Secuencia , Simportadores/biosíntesis , Simportadores/genética , Tannerella forsythia/genética , Tannerella forsythia/crecimiento & desarrollo
6.
Mol Oral Microbiol ; 39(2): 40-46, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37459655

RESUMEN

The oral organism Tannerella forsythia is auxotrophic for peptidoglycan amino sugar N-acetylmuramic acid (MurNAc). It survives in the oral cavity by scavenging MurNAc- and MurNAc-linked peptidoglycan fragments (muropeptides) secreted by co-habiting bacteria such as Fusobacterium nucleatum with which it forms synergistic biofilms. Muropeptides, MurNAc-l-Ala-d-isoGln (MDP, muramyl dipeptide) and d-γ-glutamyl-meso-DAP (iE-DAP dipeptide), are strong immunostimulatory molecules that activate nucleotide oligomerization domain (NOD)-like innate immune receptors and induce the expression of inflammatory cytokines and antimicrobial peptides. In this study, we utilized an in vitro T. forsythia-F. nucleatum co-culture model to determine if T. forsythia can selectively scavenge NOD ligands from the environment and impact NOD-mediated inflammation. The results showed that NOD-stimulatory molecules were secreted by F. nucleatum in the spent culture broth, which subsequently induced cytokine and antimicrobial peptide expression in oral epithelial cells. In the spent broth from T. forsythia-F. nucleatum co-cultures, the NOD-stimulatory activity was significantly reduced. These data indicated that F. nucleatum releases NOD2-stimulatory muropeptides in the environment, and T. forsythia can effectively scavenge the muropeptides released by co-habiting bacteria to dampen NOD-mediated host responses. This proof-of-principle study demonstrated that peptidoglycan scavenging by T. forsythia can impact the innate immunity of oral epithelium by dampening NOD activation.


Asunto(s)
Fusobacterium nucleatum , Tannerella forsythia , Tannerella forsythia/metabolismo , Fusobacterium nucleatum/fisiología , Peptidoglicano , Boca , Células Epiteliales/metabolismo , Citocinas/metabolismo
7.
Infect Immun ; 80(7): 2436-43, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22547549

RESUMEN

Tannerella forsythia is strongly associated with chronic periodontitis, an inflammatory disease of the tooth-supporting tissues, leading to tooth loss. Fusobacterium nucleatum, an opportunistic pathogen, is thought to promote dental plaque formation by serving as a bridge bacterium between early- and late-colonizing species of the oral cavity. Previous studies have shown that F. nucleatum species synergize with T. forsythia during biofilm formation and pathogenesis. In the present study, we showed that coinfection of F. nucleatum and T. forsythia is more potent than infection with either species alone in inducing NF-κB activity and proinflammatory cytokine secretion in monocytic cells and primary murine macrophages. Moreover, in a murine model of periodontitis, mixed infection with the two species induces synergistic alveolar bone loss, characterized by bone loss which is greater than the additive alveolar bone losses induced by each species alone. Further, in comparison to the single-species infection, mixed infection caused significantly increased inflammatory cell infiltration in the gingivae and osteoclastic activity in the jaw bones. These data show that F. nucleatum subspecies and T. forsythia synergistically stimulate the host immune response and induce alveolar bone loss in a murine experimental periodontitis model.


Asunto(s)
Pérdida de Hueso Alveolar/microbiología , Pérdida de Hueso Alveolar/patología , Bacteroidetes/patogenicidad , Fusobacterium nucleatum/patogenicidad , Infecciones por Bacterias Gramnegativas/patología , Periodontitis/microbiología , Periodontitis/patología , Animales , Coinfección/microbiología , Coinfección/patología , Modelos Animales de Enfermedad , Femenino , Infecciones por Bacterias Gramnegativas/microbiología , Ratones , Ratones Endogámicos BALB C
8.
Pathog Dis ; 80(1)2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35404415

RESUMEN

Tannerella forsythia is strongly implicated in the development of periodontitis, an inflammatory disease that destroys the bone and soft tissues supporting the tooth.  To date, the knowledge of the virulence attributes of T. forsythia species has mainly come from studies with a laboratory adapted strain (ATCC 43037). In this study, we focused on two T. forsythia clinical isolates, UB4 and UB20, in relation to their ability to activate macrophages. We found that these clinical isolates differentially induced proinflammatory cytokine expression in macrophages. Prominently, the expression of the chemokine protein IP-10 (CXCL10) was highly induced by UB20 as compared to UB4 and the laboratory strain ATCC 43037. Our study focused on the lipopolysaccharide component (LPS) of these strains and found that UB20 expressed a smooth-type LPS, unlike UB4 and ATCC 43037 each of which expressed a rough-type LPS. The LPS from UB20, via activation of TLR4, was found to be a highly potent inducer of IP-10 expression via signaling through STAT1 (signal transducer and activator of transcription-1). These data suggest that pathogenicity of T. forsythia species could be strain dependent and the LPS heterogeneity associated with the clinical strains might be responsible for their pathogenic potential and severity of periodontitis.


Asunto(s)
Periodontitis , Tannerella forsythia , Quimiocina CXCL10/genética , Humanos , Interferón gamma , Lipopolisacáridos , Macrófagos
9.
ACS Infect Dis ; 8(9): 1831-1838, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-35924866

RESUMEN

The human oral microbiome is the second largest microbial community in humans, harboring over 700 bacterial species, which aid in digestion and protect from growth of disease-causing pathogens. One such oral pathogen, Tannerella forsythia, along with other species, contributes to the pathogenesis of periodontitis. T. forsythia is unable to produce its own N-acetylmuramic acid (NAM) sugar, essential for peptidoglycan biosynthesis and therefore must scavenge NAM from other species with which it cohabitates. Here, we explore the recycling potential of T. forsythia for NAM uptake with a bioorthogonal modification into its peptidoglycan, allowing for click-chemistry-based visualization of the cell wall structure. Additionally, we identified NAM recycling enzyme homologues in T. forsythia that are similar to the enzymes found in Pseudomonas putida. These homologues were then genetically transformed into a laboratory safe Escherichia coli strain, resulting in the efficient incorporation of unnatural NAM analogues into the peptidoglycan backbone and its visualization, alone or in the presence of human macrophages. This strain will be useful in further studies to probe NAM recycling and peptidoglycan scavenging pathways of T. forsythia and other cohabiting bacteria.


Asunto(s)
Peptidoglicano , Pseudomonas putida , Pared Celular/química , Escherichia coli/metabolismo , Humanos , Ácidos Murámicos , Pseudomonas putida/genética , Tannerella forsythia/metabolismo
10.
Mol Oral Microbiol ; 37(2): 42-52, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34958712

RESUMEN

Recent epidemiological studies have shown that inflammatory bowel disease is associated with periodontal disease. The oral-gut microbiota axis is a potential mechanism intersecting the two diseases. Porphyromonas gingivalis is currently considered a keystone oral pathogen involved in periodontal disease pathogenesis and disease progression. Recent studies have shown that oral ingestion of P. gingivalis leads to intestinal inflammation. However, the molecular underpinnings of P. gingivalis-mediated gut inflammation have remained elusive. In this study, we show that the oral administration of P. gingivalis indeed leads to ileal inflammation and alteration in gut microbiota with significant reduction in bacterial alpha diversity despite the absence of P. gingivalis in the lower gastrointestinal tract. Utilizing an antibiotic-conditioned mouse model, cecal microbiota transfer experiments were performed to demonstrate that P. gingivalis-induced dysbiotic gut microbiota is sufficient to reproduce gut pathology. Furthermore, we observed a significant expansion in small intestinal lamina propria IL9+ CD4+ T cells, which was negatively correlated with both bacterial and fungal alpha diversity, signifying that P. gingivalis-mediated intestinal inflammation may be due to the subsequent loss of gut microbial diversity. Finally, we detected changes in gene expression related to gut epithelial barrier function, showing the potential downstream effect of intestinal IL9+ CD4+ T-cell induction. This study for the first time showed the mechanism behind P. gingivalis-mediated intestinal inflammation where P. gingivalis indirectly induces intestinal IL9+ CD4+ T cells and inflammation by altering the gut microbiota. Understanding the mechanism of P. gingivalis-mediated intestinal inflammation may lead to the development of novel therapeutic approaches to alleviate the morbidity from inflammatory bowel disease patients with periodontal disease.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Enfermedades Periodontales , Animales , Linfocitos T CD4-Positivos , Humanos , Inflamación/patología , Interleucina-9 , Ratones , Enfermedades Periodontales/microbiología , Porphyromonas gingivalis/genética , Linfocitos T
11.
Infect Immun ; 79(1): 393-401, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21078857

RESUMEN

Tannerella forsythia is a Gram-negative oral anaerobe which contributes to the development of periodontitis, an inflammatory disease of the tooth-supporting tissues leading to tooth loss. The mechanisms by which this bacterium colonizes the oral cavity are poorly understood. The bacterium has been shown to express two distinct sialidases, namely, SiaHI and NanH, with the latter being the major sialidase. Bacterial sialidases can play roles in pathogenesis by cleaving sialic acids on host glycoproteins, destroying their integrity, and/or unmasking hidden epitopes on host surfaces for colonization. In the present study, we investigated the roles of the SiaHI and NanH sialidases by generating and characterizing specific deletion mutants. Our results showed that the NanH deficiency resulted in a total loss of sialidase activity associated with the outer-membrane and secreted fractions. On the other hand, the SiaHI deficiency resulted in only a slight reduction in the total sialidase activity, with no significant differences in the levels of sialidase activity in the outer membrane or secreted fractions compared to that in the wild-type strain. The results demonstrated that NanH is both surface localized and secreted. The NanH-deficient mutant but not the SiaHI-deficient mutant was significantly attenuated in epithelial cell binding and invasion abilities compared to the wild-type strain. Moreover, the NanH-deficient mutant alone was impaired in cleaving surface sialic acids on epithelial cells. Thus, our study suggests that NanH sialidase might play roles in bacterial colonization by exposing sialic acid-hidden epitopes on epithelial cells.


Asunto(s)
Adhesión Bacteriana/fisiología , Proteínas Bacterianas/metabolismo , Células Epiteliales/microbiología , Bacterias Anaerobias Gramnegativas/enzimología , Neuraminidasa/metabolismo , Proteínas Bacterianas/genética , Línea Celular , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos
12.
Microbiology (Reading) ; 157(Pt 11): 3195-3202, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21885482

RESUMEN

The major bacterial pathogens associated with periodontitis include Tannerella forsythia. We previously discovered that sialic acid stimulates biofilm growth of T. forsythia, and that sialidase activity is key to utilization of sialoconjugate sugars and is involved in host-pathogen interactions in vitro. The aim of this work was to assess the influence of the NanH sialidase on initial biofilm adhesion and growth in experiments where the only source of sialic acid was sialoglycoproteins or human oral secretions. After showing that T. forsythia can utilize sialoglycoproteins for biofilm growth, we showed that growth and initial adhesion with sialylated mucin and fetuin were inhibited two- to threefold by the sialidase inhibitor oseltamivir. A similar reduction (three- to fourfold) was observed with a nanH mutant compared with the wild-type. Importantly, these data were replicated using clinically relevant serum and saliva samples as substrates. In addition, the ability of the nanH mutant to form biofilms on glycoprotein-coated surfaces could be restored by the addition of purified NanH, which we show is able to cleave sialic acid from the model glycoprotein fetuin and, much less efficiently, 9-O-acetylated bovine submaxillary mucin. These data show for the first time that glycoprotein-associated sialic acid is likely to be a key in vivo nutrient source for T. forsythia when growing in a biofilm, and suggest that sialidase inhibitors might be useful adjuncts in periodontal therapy.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteroidetes/metabolismo , Biopelículas/crecimiento & desarrollo , Glicoproteínas/metabolismo , Neuraminidasa/metabolismo , Adulto , Animales , Proteínas Bacterianas/genética , Bacteroidetes/enzimología , Bacteroidetes/genética , Bacteroidetes/crecimiento & desarrollo , Bovinos , Fetuínas/metabolismo , Humanos , Mucinas/metabolismo , Neuraminidasa/antagonistas & inhibidores , Oseltamivir/farmacología , Saliva/química
13.
Front Physiol ; 12: 722859, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34594237

RESUMEN

Periodontitis is a bacterially-induced inflammatory disease that leads to tooth loss. It results from the damaging effects of a dysregulated immune response, mediated largely by neutrophils, macrophages, T cells and B cells, on the tooth-supporting tissues including the alveolar bone. Specifically, infiltrating B cells at inflamed gingival sites with an ability to secrete RANKL and inflammatory cytokines are thought to play roles in alveolar bone resorption. However, the direct contribution of B cells in alveolar bone resorption has not been fully appreciated. In this study we sought to define the contribution of RANKL expressing B cells in periodontitis by employing a mouse model of pathogen-induced periodontitis that used conditional knockout mice with B cell-targeted RANKL deletion. Briefly, alveolar bone loss was assessed in the wild-type, B-cell deficient (Jh), or B-cell-RANKL deleted (RANKLΔB) mice orally infected with the periodontal pathogen Tannerella forsythia. The RANKLΔB mice were obtained by crossing Cd19-Cre knock-in mice with mice homozygous for conditional RANKL-flox allele (RANKLflox/flox). The alveolar bone resorption was determined by morphometric analysis and osteoclastic activity of the jaw bone. In addition, the bone resorptive potential of the activated effector B cells was assessed ex vivo. The data showed that the RANKL producing B cells increased significantly in the T. forsythia-infected wild-type mice compared to the sham-infected mice. Moreover, T. forsythia-infection induced higher alveolar bone loss in the wild-type and RANKLflox/flox mice compared to infection either in the B cell deficient (Jh) or the B-cell specific RANKL deletion (RANKLΔB) mice. These data established that the oral-pathogen activated B cells contribute significantly to alveolar bone resorption via RANKL production.

14.
Methods Mol Biol ; 2210: 135-142, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32815134

RESUMEN

The objective of this chapter is to provide a detailed purification protocol for the surface-layer (S-layer) glycoproteins of the periodontal pathogen Tannerella forsythia. The procedure involves detergent based solubilization of the bacterial S-layer followed by cesium chloride gradient centrifugation and gel permeation chromatography. The protocol is suitable for the isolation of S-layer glycoproteins from T. forsythia strains with diverse O-glycan structures, and aid in understanding the biochemical basis and the role of protein O-glycosylation in bacterial pathogenesis.


Asunto(s)
Proteínas Bacterianas/aislamiento & purificación , Cromatografía en Gel/métodos , Glicoproteínas de Membrana/aislamiento & purificación , Tannerella forsythia/química , Proteínas Bacterianas/química , Centrifugación/métodos , Glicosilación , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Glicoproteínas de Membrana/química , Periodontitis/microbiología , Solubilidad
15.
Infect Immun ; 76(1): 198-205, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17967853

RESUMEN

Tannerella forsythia is a gram-negative anaerobe strongly associated with chronic human periodontitis. This bacterium expresses a cell surface-associated and secreted protein, designated BspA, which has been recognized as an important virulence factor. The BspA protein belongs to the leucine-rich repeat (LRR) and bacterial immunoglobulin-like protein families. BspA is, moreover, a multifunctional protein which interacts with a variety of host cells, including monocytes which appear to respond to BspA through Toll-like receptor (TLR) signaling. Since gingival epithelium forms a barrier against periodontal pathogens, this study was undertaken to determine if gingival epithelial cells respond to BspA challenge and if TLRs play any role in BspA recognition. This study was also directed towards identifying the BspA domains responsible for cellular activation. We provide direct evidence for BspA binding to TLR2 and demonstrate that the release of the chemokine interleukin-8 from human gingival epithelial cells by BspA is TLR2 dependent. Furthermore, the LRR domain of BspA is involved in activation of TLR2, while TLR1 serves as a signaling partner. Thus, our findings suggest that BspA is an important modulator of host innate immune responses through activation of TLR2 in cooperation with TLR1.


Asunto(s)
Proteínas Bacterianas/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Encía/citología , Interleucina-8/genética , Proteínas de la Membrana/farmacología , Receptor Toll-Like 2/metabolismo , Línea Celular , Bacterias Anaerobias Gramnegativas/metabolismo , Humanos , Interleucina-8/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptor Toll-Like 1/metabolismo , Receptor Toll-Like 6/metabolismo
16.
Photodiagnosis Photodyn Ther ; 23: 18-24, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29753881

RESUMEN

BACKGROUND: Porphyromonas gingivalis and Treponema denticola are both principally implicated in the incidence of both periodontal disease and peri-implantitis. Recent studies have demonstrated that these bacteria exhibit symbiotic growth in vitro and a synergistic virulence in co-infection of animal models. Found at varying depths throughout the biofilm, these bacteria present a significant challenge to traditional antimicrobial treatment modalities. Antimicrobial photodynamic therapy (aPDT) has yielded high success against bacterial biofilms, namely those found in the oral cavity. Data on the use of aPDT against these particular periodontal pathogens is, however, scarce. Here, we studied the qualitative killing efficacy and depth of drug and laser penetration into defined P. gingivalis and T. denticola biofilms. METHODS: P. gingivalis and T. denticola were incubated under anaerobic (10%CO2, 10%H2, 80%N2) conditions for two days in diluted TSB with PBS (TYGVS for T. denticola maintenance) to elicit biofilm growth on coverslip-modified polystyrene dishes. Treated biofilms were exposed to a purpurin-based sensitizer (25 µg/mL in DMSO) for 30 min, and then aPDT was carried out using a diode laser at 664 nm. Light doses of 15 and 45 J/cm2 were used. All biofilms were then exposed to Filmtracer™ LIVE/DEAD® Biofilm Viability Kit (Cat No. L10316). Qualitative analysis was performed using a Zeiss LSM 510 Meta NLO Confocal Microscope with attached Zeiss Axioimager Z1 and Axiovert 200 M for visual data collection, and images were processed using the ZEN Digital Imaging for Light Microscopy software suite. Analysis was performed in 2 × 3 stacks to assess the entire depth of both the biofilm and presumed drug/laser penetration. RESULTS: Initial planktonic studies confirmed that the bacteria in question were present in the grown cultures and susceptible to aPDT exposure. Biofilm control groups were found to have significant levels of surviving bacterial colonies. Both treatment groups featured complete bacterial kill throughout the entirety of the biofilm (average: 23.17 µm; range: 18.13-27.20 µm). CONCLUSIONS: The efficacy of the purpurin-based PS and aPDT is demonstrated to be effective at both high and low light doses. Bacterial kill was fully efficacious at each visualized biofilm layer (1.01 µm/z-level). This study serves as a proof of concept for future studies that must consider appropriate treatment parameters, including the amount of applied PS, and laser dose. These findings indicate that aPDT is a method that can be used to eliminate microorganisms associated with biofilms implicated in the etiology of peri-implantitis and periodontitis at large.


Asunto(s)
Antraquinonas/farmacología , Biopelículas/efectos de los fármacos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Porphyromonas gingivalis/efectos de los fármacos , Treponema denticola/efectos de los fármacos , Láseres de Semiconductores , Viabilidad Microbiana , Imagen Óptica , Plancton/efectos de los fármacos
17.
PLoS One ; 12(3): e0173394, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28264048

RESUMEN

The oral pathogen Tannerella forsythia is implicated in the development of periodontitis, a common inflammatory disease that leads to the destruction of the gum and tooth supporting tissues, often leading to tooth loss. T. forsythia is a unique Gram-negative organism endowed with an elaborate protein O-glycosylation system that allows the bacterium to express a glycosylated surface (S)-layer comprising two high molecular weight glycoproteins modified with O-linked oligosaccharides. The T. forsythia S-layer has been implicated in the modulation of cytokine responses of antigen presenting cells, such as macrophages, that play a significant role during inflammation associated with periodontitis. The macrophage-inducible C-type lectin receptor (Mincle) is an FcRγ-coupled pathogen recognition receptor that recognizes a wide variety of sugar containing ligands from fungal and bacterial pathogens. In this study, we aimed to determine if Mincle might be involved in the recognition of T. forsythia S-layer and modulation of cytokine response of macrophages against the bacterium. Binding studies using recombinant Mincle-Fc fusion protein indicated a specific Ca2+-dependent binding of Mincle to T. forsythia S-layer. Subsequent experiments with Mincle-expressing and Mincle-knockdown macrophages revealed a role for Mincle/S-layer interaction in the induction of both pro- and anti-inflammatory cytokine secretion in macrophages stimulated with T. forsythia as well as its S-layer. Together, these studies revealed Mincle as an important macrophage receptor involved in the modulation of cytokine responses of macrophages against T. forsythia, and thus may play a critical role in orchestrating the host immune response against the bacterium.


Asunto(s)
Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Periodontitis/microbiología , Tannerella forsythia/inmunología , Tannerella forsythia/metabolismo , Diferenciación Celular , Línea Celular , Citocinas/metabolismo , Glicosilación , Humanos , Activación de Macrófagos/inmunología , Macrófagos/citología , Macrófagos/inmunología , Periodontitis/genética , Periodontitis/inmunología , Fagocitosis/inmunología , Unión Proteica , ARN Interferente Pequeño/genética , Tannerella forsythia/patogenicidad
18.
Front Microbiol ; 8: 648, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28446907

RESUMEN

Tannerella forsythia is a Gram-negative oral anaerobe associated with periodontitis. This bacterium is auxotrophic for the peptidoglycan amino sugar N-acetylmuramic (MurNAc) and likely relies on scavenging peptidoglycan fragments (muropeptides) released by cohabiting bacteria during their cell wall recycling. Many Gram-negative bacteria utilize an inner membrane permease, AmpG, to transport peptidoglycan fragments into their cytoplasm. In the T. forsythia genome, the Tanf_08365 ORF has been identified as a homolog of AmpG permease. In order to confirm the functionality of Tanf_08365, a reporter system in an Escherichia coli host was generated that could detect AmpG-dependent accumulation of cytosolic muropeptides via a muropeptide-inducible ß-lactamase reporter gene. In trans complementation of this reporter strain with a Tanf_08365 containing plasmid caused significant induction of ß-lactamase activity compared to that with an empty plasmid control. These data indicated that Tanf_08365 acted as a functional muropeptide permease causing accumulation of muropeptides in E. coli and thus suggested that it is a permease involved in muropeptide scavenging in T. forsythia. Furthermore, we showed that the promoter regulating the expression of Tanf_08365 was activated significantly by a hybrid two-component system regulatory protein, GppX. We also showed that compared to the parental T. forsythia strain a mutant lacking GppX in which the expression of AmpG was reduced significantly attenuated in utilizing free muropeptides. In summary, we have uncovered the mechanism by which this nutritionally fastidious microbe accesses released muropeptides in its environment, opening up the possibility of targeting this activity to reduce its numbers in periodontitis patients with potential benefits in the treatment of disease.

19.
FEMS Microbiol Lett ; 264(1): 15-21, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17020544

RESUMEN

Porphyromonas gingivalis is recognized as one of the major periodontal pathogens in subgingival plaque, which is implicated in the progression of chronic periodontal disease. We analyzed the role of upsA in P. gingivalis 381 and its uspA-deficient mutant CW301 under various stress conditions. In general, the uspA mutant was less tolerant to a variety of environmental stresses relative to the parental strain. In addition, gene expression of uspA is upregulated during biofilm formation. Biofilm formation of the uspA mutant was also less than that of strain 381. In conclusion, the uspA gene affecting the stress responses of P. gingivalis is required for optimal biofilm formation.


Asunto(s)
Proteínas Bacterianas/fisiología , Biopelículas/crecimiento & desarrollo , Proteínas de Choque Térmico/fisiología , Porphyromonas gingivalis/fisiología , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Silenciador del Gen , Proteínas de Choque Térmico/genética , Calor , Pruebas de Sensibilidad Microbiana , Estrés Oxidativo , Porphyromonas gingivalis/efectos de los fármacos , Porphyromonas gingivalis/metabolismo , Tetraciclina/farmacología
20.
Photodiagnosis Photodyn Ther ; 13: 22-28, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26617192

RESUMEN

BACKGROUND: Dental implants are commonly used today for the treatment of partially and fully edentulous patients. Despite the high success rate they are not resistant to complications and failure due to a variety of problems including peri-implantitis or peri-mucositis due to bacterial biofilm formation on the implant surface. The use of non-surgical and surgical treatment procedure to promote healing in cases with peri-implantitis have limited efficacy. Here we studied the ability of photodynamic therapy to destroy a known bacterial pathogen and the extracellular matrix architecture of biofilm attached to titanium plates and germanium prisms. METHODS: Titanium plates or germanium prisms were incubated for 24h with Fusobacterium nucleatum a fusiform, gram-negative bacterium was used to enable biofilm formation. Photodynamic therapy was carried out by incubating the biofilm samples on each substrata with porfimer sodium. Treatment was carried out using a diode laser at 630nm, 150mW/cm(2) for light doses ranging from 25-100J/cm(2). Evaluation of killing efficacy was done by counting colony forming units compared to controls. Multiple attenuated internal reflection-infrared spectroscopy (MAIR-IR) and SEM were used to analyze the samples pre and post PDT for validation. RESULTS: F. nucleatum was significantly reduced in a dose dependent manner by treatment with PDT. Changes in biofilm components and strength of bioadhesion were examined with MAIR-IR following jet impingement using calibrated water jets. SEM demonstrates significant morphological alterations in the bacteria, consistent with damage associated with exposure to reactive oxygen species. CONCLUSION: The results are indicative that aPDT is a method that can be used to eradicate micro-organisms associated with biofilm in peri-implantitis on relevant substrata. Data shows that the slime layer of the biofilm is removed and that further methods need to be employed to completely remove weakened or destroyed biofilm matrix components. Reactive oxygen species (ROS) mediated oxidative damage results in morphologic changes as a consequence of changes in cell membrane integrity.


Asunto(s)
Biopelículas/efectos de los fármacos , Implantes Dentales/microbiología , Éter de Dihematoporfirina/administración & dosificación , Matriz Extracelular/efectos de los fármacos , Fusobacterias/efectos de los fármacos , Fotoquimioterapia/métodos , Biopelículas/crecimiento & desarrollo , Matriz Extracelular/patología , Matriz Extracelular/efectos de la radiación , Fusobacterias/fisiología , Fusobacterias/efectos de la radiación , Fármacos Fotosensibilizantes , Esterilización/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA