Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 834
Filtrar
1.
Biochem Biophys Res Commun ; 707: 149783, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38493746

RESUMEN

Ingestion of Porphyromonas gingivalis, a periodontal pathogen, disrupts the intestinal barrier in mice. However, the involvement of outer membrane vesicles (OMVs) secreted from P. gingivalis in the destruction of the intestinal barrier remains unclear. In this study, we tested the hypothesis that OMVs carrying gingipains, the major cysteine proteases produced by P. gingivalis, affects the intestinal barrier function. OMVs increased the permeability of the Caco-2 cell monolayer, a human intestinal epithelial cell line, accompanied by degradation of the tight junction protein occludin. In contrast, OMVs prepared from mutant strains devoid of gingipains failed to induce intestinal barrier dysfunction or occludin degradation in Caco-2 cells. A close histological examination revealed the intracellular localization of gingipain-carrying OMVs. Gingipain activity was detected in the cytosolic fraction of Caco-2 cells after incubation with OMVs. These results suggest that gingipains were internalized into intestinal cells through OMVs and transported into the cytosol, where they then directly degraded occludin from the cytosolic side. Thus, P. gingivalis OMVs might destroy the intestinal barrier and induce systemic inflammation via OMV itself or intestinal substances leaked into blood vessels, causing various diseases.


Asunto(s)
Adhesinas Bacterianas , Porphyromonas gingivalis , Animales , Ratones , Humanos , Cisteína-Endopeptidasas Gingipaínas/metabolismo , Células CACO-2 , Porphyromonas gingivalis/fisiología , Citosol/metabolismo , Ocludina/metabolismo , Adhesinas Bacterianas/metabolismo
2.
Theranostics ; 14(4): 1781-1793, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38389835

RESUMEN

Candida albicans and Porphyromonas gingivalis are prevalent in the subgingival area where the frequency of fungal colonization increases with periodontal disease. Candida's transition to a pathogenic state and its interaction with P. gingivalis exacerbate periodontal disease severity. However, current treatments for these infections differ, and combined therapy remains unexplored. This work is based on an antimicrobial peptide that is therapeutic and induces a color change in a nanoparticle reporter. Methods: We built and characterized two enzyme-activatable prodrugs to treat and detect C. albicans and P. gingivalis via the controlled release of the antimicrobial peptide. The zwitterionic prodrug quenches the antimicrobial peptide's activity until activation by a protease inherent to the pathogens (SAP9 for C. albicans and RgpB for P. gingivalis). The toxicity of the intact prodrugs was evaluated against fungal, bacterial, and mammalian cells. Therapeutic efficacy was assessed through microscopy, disk diffusion, and viability assays, comparing the prodrug to the antimicrobial peptide alone. Finally, we developed a colorimetric detection system based on the aggregation of plasmonic nanoparticles. Results: The intact prodrugs showed negligible toxicity to cells absent a protease trigger. The therapeutic impact of the prodrugs was comparable to that of the antimicrobial peptide alone, with a minimum inhibitory concentration of 3.1 - 16 µg/mL. The enzymatic detection system returned a detection limit of 10 nM with gold nanoparticles and 3 nM with silver nanoparticles. Conclusion: This approach offers a convenient and selective protease sensing and protease-induced treatment mechanism based on bioinspired antimicrobial peptides.


Asunto(s)
Nanopartículas del Metal , Enfermedades Periodontales , Profármacos , Animales , Porphyromonas gingivalis/fisiología , Candida albicans/fisiología , Péptido Hidrolasas , Péptidos Antimicrobianos , Profármacos/farmacología , Preparaciones de Acción Retardada , Oro , Plata , Endopeptidasas , Mamíferos
3.
J Dent Res ; 103(2): 197-207, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38185909

RESUMEN

Periodontitis has been emphasized as a risk factor of insulin resistance-related systemic diseases. Accumulating evidence has suggested a possible "oral-gut axis" linking oral infection and extraoral diseases, but it remains unclear whether periodontal pathogens can survive the barriers of the digestive tract and how they play their pathogenic roles. The present study established a periodontitis mouse model through oral ligature plus Porphyromonas gingivalis inoculation and demonstrated that periodontitis aggravated diet-induced obesity and insulin resistance, while also causing P. gingivalis enrichment in the intestine. Metabolic labeling strategy validated that P. gingivalis could translocate to the gastrointestinal tract in a viable state. Oral administration of living P. gingivalis elicited insulin resistance, while administration of pasteurized P. gingivalis had no such effect. Combination analysis of metagenome sequencing and nontargeted metabolomics suggested that the tryptophan metabolism pathway, specifically indole and its derivatives, was involved in the pathogenesis of insulin resistance caused by oral administration of living P. gingivalis. Moreover, liquid chromatography-high-resolution mass spectrometry analysis confirmed that the aryl hydrocarbon receptor (AhR) ligands, mainly indole acetic acid, tryptamine, and indole-3-aldehyde, were reduced in diet-induced obese mice with periodontitis, leading to inactivation of AhR signaling. Supplementation with Ficz (6-formylindolo (3,2-b) carbazole), an AhR agonist, alleviated periodontitis-associated insulin resistance, in which the restoration of gut barrier function might play an important role. Collectively, these findings reveal that the oral-gut translocation of viable P. gingivalis works as a fuel linking periodontitis and insulin resistance, in which reduction of AhR ligands and inactivation of AhR signaling are involved. This study provides novel insight into the role of the oral-gut axis in the pathogenesis of periodontitis-associated comorbidities.


Asunto(s)
Resistencia a la Insulina , Periodontitis , Ratones , Animales , Porphyromonas gingivalis/fisiología , Ratones Endogámicos C57BL , Periodontitis/metabolismo , Modelos Animales de Enfermedad
4.
Exp Biol Med (Maywood) ; 248(23): 2227-2236, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38073544

RESUMEN

Antibiotics can kill bacteria, but their continued use can easily lead to drug resistance, particularly the main pathogenic bacteria of periodontitis, Porphyromonas gingivalis. However, to avoid drug resistance, carbon quantum dots (CDs) have great potential as a bioactive material in antimicrobial therapy. Herein, we use ornidazole as raw material to prepare CDs of different sizes by microwave irradiation and screen CDs with fluorescence and bacteriostatic properties. The inhibition experiments and live/dead assays of P. gingivalis exhibited outstanding antibacterial effects. This research aimed to develop nano-level antibacterial active materials that also have fluorescence traceability. This study offers a different method for the development of multifunctional CDs, provides valuable strategies for the treatment of diseases associated with P. gingivalis, and predicts great application prospects in the field of biomedicine.


Asunto(s)
Puntos Cuánticos , Porphyromonas gingivalis/fisiología , Carbono/farmacología , Antibacterianos/farmacología , Colorantes
5.
Appl Environ Microbiol ; 89(10): e0108123, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37768099

RESUMEN

Biofilms are complex polymicrobial communities which are often associated with human infections such as the oral disease periodontitis. Studying these complex communities under controlled conditions requires in vitro biofilm model systems that mimic the natural environment as close as possible. This study established a multispecies periodontal model in the drip flow biofilm reactor in order to mimic the continuous flow of nutrients at the air-liquid interface in the oral cavity. The design is engineered to enable real-time characterization. A community of five bacteria, Streptococcus gordonii-GFPmut3*, Streptococcus oralis-GFPmut3*, Streptococcus sanguinis-pVMCherry, Fusobacterium nucleatum, and Porphyromonas gingivalis-SNAP26 is visualized using two distinct fluorescent proteins and the SNAP-tag. The biofilm in the reactor develops into a heterogeneous, spatially uniform, dense, and metabolically active biofilm with relative cell abundances similar to those in a healthy individual. Metabolic activity, structural features, and bacterial composition of the biofilm remain stable from 3 to 6 days. As a proof of concept for our periodontal model, the 3 days developed biofilm is exposed to a prebiotic treatment with L-arginine. Multifaceted effects of L-arginine on the oral biofilm were validated by this model setup. L-arginine showed to inhibit growth and incorporation of the pathogenic species and to reduce biofilm thickness and volume. Additionally, L-arginine is metabolized by Streptococcus gordonii-GFPmut3* and Streptococcus sanguinis-pVMCherry, producing high levels of ornithine and ammonium in the biofilm. In conclusion, our drip flow reactor setup is promising in studying spatiotemporal behavior of a multispecies periodontal community.ImportancePeriodontitis is a multifactorial chronic inflammatory disease in the oral cavity associated with the accumulation of microorganisms in a biofilm. Not the presence of the biofilm as such, but changes in the microbiota (i.e., dysbiosis) drive the development of periodontitis, resulting in the destruction of tooth-supporting tissues. In this respect, novel treatment approaches focus on maintaining the health-associated homeostasis of the resident oral microbiota. To get insight in dynamic biofilm responses, our research presents the establishment of a periodontal biofilm model including Streptococcus gordonii, Streptococcus oralis, Streptococcus sanguinis, Fusobacterium nucleatum, and Porphyromonas gingivalis. The added value of the model setup is the combination of simulating continuously changing natural mouth conditions with spatiotemporal biofilm profiling using non-destructive characterization tools. These applications are limited for periodontal biofilm research and would contribute in understanding treatment mechanisms, short- or long-term exposure effects, the adaptation potential of the biofilm and thus treatment strategies.


Asunto(s)
Bacterias , Periodontitis , Humanos , Streptococcus gordonii/fisiología , Fusobacterium nucleatum , Streptococcus sanguis , Streptococcus oralis , Biopelículas , Arginina/metabolismo , Porphyromonas gingivalis/fisiología
6.
Int. j. morphol ; 41(2): 431-436, abr. 2023. ilus, tab
Artículo en Español | LILACS | ID: biblio-1440308

RESUMEN

La enfermedad periodontal es una de las principales causas de pérdida dentaria. Clínicamente, esta patología, mediada por la desregulación del sistema inmune producto de una disbiosis ocurrida en el surco gingival, inicia con la inflamación de la encía y evoluciona con el daño irreversible de los tejidos que rodean el diente. El hueso alveolar es uno de los tejidos afectados esta patología, esto debido a la activación de osteoclastos por la sobreexpresión de la proteína RANKL en el huésped. El propósito de este trabajo es determinar el nivel de sobreexpresión de RANKL, en un modelo de células tumorales U2OS, frente a la infección con Porphyromonas gingivalis y Prevotella intermedia. Para identificar el nivel de RANKL, se definieron cuatro grupos: Un grupo control, no tratado; Grupo PG, tratado con P. gingivalis; Grupo PI, tratado con P. Intermedia; y un grupo PG+PI, tratado con ambas bacterias. El nivel relativo de la proteína RANKL fue determinado en el sobrenadante y en los extractos celulares de manera independiente, mediante la técnica Western blot. En sobrenadantes, el grupo PG mostró mayores niveles de RANKL comparados con PI (p < 0,05). En extractos celulares los niveles fueron mayores en el grupo PG+PI (p < 0,05). El grupo PI mostró los niveles más bajos de RANKL. La infección polimicrobiana resulta en una mayor expresión de RANKL en células tumorales U2OS, mientras que frente a la infección P. gingivalis, se observó mayor cantidad de RANKL soluble.


SUMMARY: Periodontal disease is one of the main causes of tooth loss. Clinically, this pathology, mediated by the deregulation of the immune system due to a dysbiosis occurred in the gingival sulcus, begins with the inflammation of the gum and evolves with the irreversible damage of the tissues that surround the tooth. Alveolar bone is one of the most affected tissues by this disease, due to the activation of osteoclasts by the upregulation of RANKL in the host. The aim of this study is to determine the increase of RANKL, in a U2OS tumor cells model, inoculated with Porphyromonas gingivalis and Prevotella intermedia. To identify the level of RANKL, four groups were defined: A control group, not treated; PG group, treated with P.gingivalis; PI group, treated with P. intermedia; and a PG+PI group, treated with both bacteria. The relative level of RANKL was determined in the supernatant and cell extracts independently, using the Western blot technique. In supernatants, the PG group showed higher RANKL levels compared to PI (p < 0.05). In cell extracts the levels were higher in the PG+PI group (p < 0.05.). The PI group showed the lowest levels of RANKL.Polymicrobial infection results in a greater expression of of soluble RANKL was observed.


Asunto(s)
Enfermedades Periodontales/microbiología , Bacterias Anaerobias/fisiología , Resorción Ósea/microbiología , Ligando RANK/metabolismo , Células Cultivadas , Western Blotting , Porphyromonas gingivalis/fisiología , Prevotella intermedia/fisiología , Línea Celular Tumoral , Electroforesis , Ligando RANK/análisis
7.
J Adv Res ; 50: 55-68, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36243399

RESUMEN

INTRODUCTION: Serum amyloid P component (SAP) regulates the innate immune system and microbial diseases. Periodontitis is an inflammatory oral disease developed by the host immune system's interaction with the dysbiotic oral microbiome, thereby SAP could play a role in periodontitis pathogenicity. OBJECTIVES: To investigate the role of SAP in oral microbiome modulation and peridontitis pathogenicity. METHODS: In this study, wildtype and SAP-knockout (KO) mice were used. Ligature-based periodontitis was developed in mice. Oral microbiome diversity was analyzed by 16 s rRNA sequencing. Macrophages and Porphyromonas gingivalis (P. gingivalis) co-culture system analyzed the effect of SAP in macrophage phagocytosis of P. gingivalis. RESULTS: The level of SAP was upregulated in the periodontitis-affected periodontium of humans and mice but not in the liver and blood circulation. Periodontal macrophages were the key source of upregulated SAP in periodontitis. SAP-KO aggravated periodontal inflammation, periodontitis, and a higher number of M1-type inflammatory macrophage infiltration in the periodontium. The oral microbiome of SAP-KO periodontitis mice was altered with a higher abundance of Porphyromonas at the genus level. SAP-KO macrophages showed compromised phagocytosis of P. gingivalis in the co-culture system. Co-culture of SAP-KO macrophages and P. gingivalis induced the C5a expression and exogenous SAP treatment nullified this effect. Exogenous recombinant SAP treatment did not affect P. gingivalis growth and opsonization. PMX205, an antagonist of C5a, treatment robustly enhanced P. gingivalis phagocytosis by SAP-KO macrophages, indicating the involvement of the C5a-C5aR signaling in the compromised P. gingivalis phagocytosis by SAP-KO macrophages. CONCLUSION: SAP deficiency aggravates periodontitis possibly via C5a-C5aR signaling-mediated defective macrophage phagocytosis of P. gingivalis. A higher abundance of P. gingivalis during SAP deficiency could promote M1 macrophage polarization and periodontitis. This finding suggests the possible protecting role of elevated levels of periodontal SAP against periodontitis progression.


Asunto(s)
Periodontitis , Porphyromonas gingivalis , Animales , Humanos , Ratones , Macrófagos/metabolismo , Ratones Noqueados , Periodontitis/metabolismo , Fagocitosis , Porphyromonas gingivalis/fisiología , Transducción de Señal , Componente Amiloide P Sérico/metabolismo
8.
ACS Nano ; 16(11): 18253-18265, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36288552

RESUMEN

Periodontitis is a chronic inflammatory disease caused by the interaction of oral microorganisms with the host immune response. Porphyromonas gingivalis (P.g.) acts as a key mediator in subverting the homeostasis of the local immune system. On the one hand, P.g. inhibits phagocytosis and the killing capacity of immune cells. On the other hand, P.g. increases selective cytokine release, which is beneficial to its further proliferation. Here, we prepared a penetrating macrophage-based nanoformulation (MZ@PNM)-encapsulating hydrogel (MZ@PNM@GCP) that responded to the periodontitis microenvironment. MZ@PNM targeted P.g. via the Toll-like receptor complex 2/1 (TLR2/1) on its macrophage-mimicking membrane, then directly killed P.g. through disruption of bacterial structural integrity by the cationic nanoparticles and intracellular release of an antibacterial drug, metronidazole (MZ). Meanwhile, MZ@PNM interrupted the specific binding of P.g. to immune cells and neutralized complement component 5a (C5a), preventing P.g. subversion of periodontal host immune response. Overall, MZ@PNM@GCP showed potent efficacy in periodontitis treatment, restoring local immune function and killing pathogenic bacteria, while exhibiting favorable biocompatibility, all of which have been demonstrated both in vivo and in vitro.


Asunto(s)
Periodontitis , Humanos , Periodontitis/tratamiento farmacológico , Periodontitis/metabolismo , Porphyromonas gingivalis/fisiología , Macrófagos/metabolismo , Citocinas
9.
Biomater Adv ; 142: 213158, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36288629

RESUMEN

Bacteria are recognized as the driving factors of periodontitis. However, excessive reactive oxygen species (ROS) can harm periodontal tissue while also causing an uncontrolled inflammatory response. Hence, eliminating excessive ROS and blocking ROS-induced abnormal inflammatory response by antioxidants are achieving remarkable results in periodontitis therapy. Moreover, influenced by the deep and irregular periodontal pockets, injectable thermo-sensitive chitosan-based hydrogels have attracted a lot of attention. This study aimed to formulate an antibacterial and antioxidant therapeutic regimen by incorporating antimicrobial peptides (Nal-P-113) and/or antioxidants (polydopamine nanoparticles, PDNPs) into chitosan-based hydrogels. The hydrogel was characterized in vitro and finally examined in rats using the experimental periodontitis model. The release kinetics showed that the hydrogel could stably release Nal-P-113 and PDNPs for up to 13 days. The scavenging activity of the hydrogel against DPPH was about 80 % and the antibacterial ratio against Streptococcus gordonii (S. gordonii), Fusobacterium nucleatum (F. nucleatum) and Porphyromonas gingivalis (P. gingivalis) was about 99 %. Importantly, it was examined that the hydrogel had the ability to prevent periodontal tissue damage. Thus, chitosan-based hydrogels may provide a basis for designing multifunctional local drug delivery biomaterials for the treatment of periodontitis.


Asunto(s)
Quitosano , Periodontitis , Ratas , Animales , Quitosano/química , Hidrogeles/química , Antioxidantes/farmacología , Especies Reactivas de Oxígeno/uso terapéutico , Periodontitis/tratamiento farmacológico , Porphyromonas gingivalis/fisiología , Antibacterianos/farmacología
10.
Cell Rep ; 40(10): 111314, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36070692

RESUMEN

Host immune response via Th17 cells against oral pathobionts is a key mediator in periodontitis development. However, where and how the Th17-type immune response is induced during the development of periodontitis is not well understood. Here, we demonstrate that gut translocation of the oral pathobiont Porphyromonas gingivalis (Pg) exacerbates oral pathobiont-induced periodontitis with enhanced Th17 cell differentiation. The oral pathobiont-responsive Th17 cells are differentiated in Peyer's patches and translocated systemically in the peripheral immune tissues. They are also capable of migrating to and accumulating in the mouth upon oral infection. Development of periodontitis via the oral pathobiont-responsive Th17 cells is regulated by the intestinal microbiome, and altering the intestinal microbiome composition with antibiotics affects the development of periodontitis. Our study highlights that pathobiont-responsive Th17 cells in the gut-mouth axis and the intestinal microbiome work together to provoke inflammatory oral diseases, including periodontitis.


Asunto(s)
Microbioma Gastrointestinal , Periodontitis , Humanos , Porphyromonas gingivalis/fisiología , Células Th17
11.
PLoS One ; 17(9): e0274523, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36103549

RESUMEN

The objective of this study was to investigate the treatment effects of non-thermal atmospheric gas plasmas (NTAP) on destruction and the recovery (or re-colonization) of Porphyromonas gingivalis (P. gingivalis) in biofilms. P. gingivalis is a well-known keystone periodontal pathogen strongly associated with periodontal diseases, especially periodontitis. P. gingivalis biofilms were formed on stainless steel coupons and treated for 1, 2, and 5 minutes by NTAP of pure argon gas and argon+oxygen gas mixture. MTT assay, colony forming unit (CFU) counting assay and confocal laser scanning microscopy (CLSM) were used to assess the destruction efficiency. In addition, the plasma treated biofilms were re-cultured in the medium supplemented with antibiotics and oxidative stress sources to determine the synergy of the NTAP with other antimicrobial agents. The results showed the plasma treatment could result in 2.7 log unit reduction in bacterial load. The recovered biofilm CFU with NTAP treatment combined with sub minimal inhibition concentration of amoxicillin was 0.33 log units less than the biofilm treated with amoxicillin alone. The recovered biofilm CFU in NTAP groups was about 2.0 log units less than that in the untreated controls under H2O2 treatment. There was approximately 1.0 log unit reduction of biofilm CFU in plasma treated biofilm compared with untreated control under paraquat treatment. The plasma treated biofilms exhibited less resistance to amoxicillin and greater susceptibility to hydrogen peroxide (H2O2) and paraquat, suggesting that NTAP may enhance biofilm susceptibility to host defense. These in vitro findings suggested that NTAP could be a novel and effective treatment method of oral biofilms that cause periodontal diseases.


Asunto(s)
Enfermedades Periodontales , Gases em Plasma , Amoxicilina/farmacología , Argón/farmacología , Biopelículas , Humanos , Peróxido de Hidrógeno/farmacología , Paraquat/farmacología , Gases em Plasma/farmacología , Porphyromonas gingivalis/fisiología
12.
Microbiol Spectr ; 10(5): e0075322, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36000865

RESUMEN

Porphyromonas gingivalis is a keystone oral pathogen that successfully manipulates the human innate immune defenses, resulting in a chronic proinflammatory state of periodontal tissues and beyond. Here, we demonstrate that secreted outer membrane vesicles (OMVs) are deployed by P. gingivalis to selectively coat and activate human neutrophils, thereby provoking degranulation without neutrophil killing. Secreted granule components with antibacterial activity, especially LL-37 and myeloperoxidase (MPO), are subsequently degraded by potent OMV-bound proteases known as gingipains, thereby ensuring bacterial survival. In contrast to neutrophils, the P. gingivalis OMVs are efficiently internalized by macrophages and epithelial cells. Importantly, we show that neutrophil coating is a conserved feature displayed by OMVs of at least one other oral pathogen, namely, Aggregatibacter actinomycetemcomitans. We conclude that P. gingivalis deploys its OMVs for a neutrophil-deceptive strategy to create a favorable inflammatory niche and escape killing. IMPORTANCE Severe periodontitis is a dysbiotic inflammatory disease that affects about 15% of the adult population, making it one of the most prevalent diseases worldwide. Importantly, periodontitis has been associated with the development of nonoral diseases, such as rheumatoid arthritis, pancreatic cancer, and Alzheimer's disease. Periodontal pathogens implicated in periodontitis can survive in the oral cavity only by avoiding the insults of neutrophils while at the same time promoting an inflamed environment where they successfully thrive. Our present findings show that outer membrane vesicles secreted by the keystone pathogen Porphyromonas gingivalis provide an effective delivery tool of virulence factors that protect the bacterium from being killed while simultaneously activating human neutrophils.


Asunto(s)
Neutrófilos , Periodontitis , Humanos , Antibacterianos , Membrana Externa Bacteriana , Cisteína-Endopeptidasas Gingipaínas , Neutrófilos/metabolismo , Periodontitis/microbiología , Peroxidasa/metabolismo , Porphyromonas gingivalis/fisiología , Factores de Virulencia/metabolismo
13.
Sci Rep ; 12(1): 14239, 2022 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-35987920

RESUMEN

Dysbiosis of the oral microbiota plays an important role in the progression of periodontitis, which is characterized by chronic inflammation and alveolar bone loss, and associated with systemic diseases. Bacterial extracellular vesicles (EVs) contain various bioactive molecules and show diverse effects on host environments depending on the bacterial species. Recently, we reported that EVs derived from Filifactor alocis, a Gram-positive periodontal pathogen, had osteoclastogenic activity. In the present study, we analysed the osteoclastogenic potency and immunostimulatory activity of EVs derived from the Gram-negative periodontal pathogens Porphyromonas gingivalis and Tannerella forsythia, the oral commensal bacterium Streptococcus oralis, and the gut probiotic strain Lactobacillus reuteri. Bacterial EVs were purified by density gradient ultracentrifugation using OptiPrep (iodixanol) reagent. EVs from P. gingivalis, T. forsythia, and S. oralis increased osteoclast differentiation and osteoclstogenic cytokine expression in osteoclast precursors, whereas EVs from L. reuteri did not. EVs from P. gingivalis, T. forsythia, and S. oralis preferentially activated Toll-like receptor 2 (TLR2) rather than TLR4 or TLR9, and induced osteoclastogenesis mainly through TLR2. The osteoclastogenic effects of EVs from P. gingivalis and T. forsythia were reduced by both lipoprotein lipase and polymyxin B, an inhibitor of lipopolysaccharide (LPS), while the osteoclastogenic effects of EVs from S. oralis were reduced by lipoprotein lipase alone. These results demonstrate that EVs from periodontal pathogens and oral commensal have osteoclastogenic activity through TLR2 activation by lipoproteins and/or LPS.


Asunto(s)
Vesículas Extracelulares , Boca , Osteoclastos , Diferenciación Celular , Vesículas Extracelulares/metabolismo , Lipopolisacáridos , Lipoproteína Lipasa , Microbiota , Boca/microbiología , Osteoclastos/metabolismo , Porphyromonas gingivalis/fisiología , Receptor Toll-Like 2 , Receptor Toll-Like 4
14.
Front Cell Infect Microbiol ; 12: 892610, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35846745

RESUMEN

Autophagy is an immune homeostasis process induced by multiple intracellular and extracellular signals. Inflammation is a protective response to harmful stimuli such as pathogen microbial infection and body tissue damage. Porphyromonas gingivalis infection elicits both autophagy and inflammation, and dysregulation of autophagy and inflammation promotes pathology. This review focuses on the interaction between autophagy and inflammation caused by Porphyromonas gingivalis infection, aiming to elaborate on the possible mechanism involved in the interaction.


Asunto(s)
Autofagia , Porphyromonas gingivalis , Autofagia/fisiología , Homeostasis , Humanos , Inflamación , Porphyromonas gingivalis/fisiología
15.
Sci Rep ; 12(1): 12161, 2022 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-35842460

RESUMEN

The aim of this study was to design and evaluate the specificity of a targeted bio-theragnostic system based on DNA-aptamer-nanographene oxide (NGO) against Porphyromonas gingivalis during antimicrobial photodynamic therapy (aPDT). Following synthesis and confirmation of NGO, the binding of selected labeled DNA-aptamer to NGO was performed and its hemolytic activity, cytotoxic effect, and release times were evaluated. The specificity of DNA-aptamer-NGO to P. gingivalis was determined. The antimicrobial effect, anti-biofilm potency, and anti-metabolic activity of aPDT were then assessed after the determination of the bacteriostatic and bactericidal concentrations of DNA-aptamer-NGO against P. gingivalis. Eventually, the apoptotic effect and anti-virulence capacity of aPDT based on DNA-aptamer-NGO were investigated. The results showed that NGO with a flaky, scale-like, and layered structure in non-cytotoxic DNA-aptamer-NGO has a continuous release in the weak-acid environment within a period of 240 h. The binding specificity of DNA-aptamer-NGO to P. gingivalis was confirmed by flow cytometry. When irradiated, non-hemolytic DNA-aptamer-NGO were photoactivated, generated ROS, and led to a significant decrease in the cell viability of P. gingivalis (P < 0.05). Also, the data indicated that DNA-aptamer-NGO-mediated aPDT led to a remarkable reduction of biofilms and metabolic activity of P. gingivalis compared to the control group (P < 0.05). In addition, the number of apoptotic cells increased slightly (P > 0.05) and the expression level of genes involved in bacterial biofilm formation and response to oxidative stress changed significantly after exposure to aPDT. It is concluded that aPDT using DNA-aptamer-NGO as a targeted bio-theragnostic system is a promising approach to detect and eliminate P. gingivalis as one of the main bacteria involved in periodontitis in periopathogenic complex in real-time and in situ.


Asunto(s)
Antiinfecciosos , Aptámeros de Nucleótidos , Fotoquimioterapia , Antibacterianos/farmacología , Aptámeros de Nucleótidos/farmacología , Biopelículas , ADN , Óxidos/farmacología , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Porphyromonas gingivalis/fisiología
16.
mBio ; 13(3): e0378721, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35491845

RESUMEN

Porphyromonas gingivalis, a keystone pathogen in periodontitis (PD), produces cysteine proteases named gingipains (RgpA, RgpB, and Kgp), which strongly affect the host immune system. The range of action of gingipains is extended by their release as components of outer membrane vesicles, which efficiently diffuse into surrounding gingival tissues. However, away from the anaerobic environment of periodontal pockets, increased oxygen levels lead to oxidation of the catalytic cysteine residues of gingipains, inactivating their proteolytic activity. In this context, the influence of catalytically inactive gingipains on periodontal tissues is of significant interest. Here, we show that proteolytically inactive RgpA induced a proinflammatory response in both gingival keratinocytes and dendritic cells. Inactive RgpA is bound to the cell surface of gingival keratinocytes in the region of lipid rafts, and using affinity chromatography, we identified RgpA-interacting proteins, including epidermal growth factor receptor (EGFR). Next, we showed that EGFR interaction with inactive RgpA stimulated the expression of inflammatory cytokines. The response was mediated via the EGFR-phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) signaling pathway, which when activated in the gingival tissue rich in dendritic cells in the proximity of the alveolar bone, may significantly contribute to bone resorption and the progress of PD. Taken together, these findings broaden our understanding of the biological role of gingipains, which in acting as proinflammatory factors in the gingival tissue, create a favorable milieu for the growth of inflammophilic pathobionts. IMPORTANCE Gingipain cysteine proteases are essential virulence factors of Porphyromonas gingivalis, an oral bacterium implicated in development of periodontitis. Gingipains diffusing from anaerobic periodontal pockets lose proteolytic activity in the oxygenated environment of gingival tissues. We found that despite the loss of activity, gingipains still elicit a strong inflammatory response, which may contribute to the progression of periodontitis and bone resorption. Moreover, we identified the host molecules utilized by the pathogen as receptors for proteolytically inactivated gingipains. The broad distribution of those receptors in human tissue suggests their involvement in systemic diseases associated with periodontal pathogens.


Asunto(s)
Resorción Ósea , Periodontitis , Adhesinas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Receptores ErbB/metabolismo , Cisteína-Endopeptidasas Gingipaínas , Humanos , Inmunidad , Bolsa Periodontal , Periodontitis/microbiología , Fosfatidilinositol 3-Quinasas/metabolismo , Porphyromonas gingivalis/fisiología
17.
Int J Mol Sci ; 23(10)2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35628542

RESUMEN

Porphyromonas gingivalis as the keystone periodontopathogen plays a critical role in the pathogenesis of periodontitis, and crucially accounts for inflammatory comorbidities such as cardiovascular disease and Alzheimer's disease. We recently identified the existence of P. gingivalis persisters and revealed the unforeseen perturbation of innate response in human gingival epithelial cells (HGECs) due to these noxious persisters. Herein, RNA sequencing revealed how P. gingivalis persisters affected the expression profile of cytokine genes and related signaling pathways in HGECs. Results showed that metronidazole-treated P. gingivalis persisters (M-PgPs) impaired the innate host defense of HGECs, in a similar fashion to P. gingivalis. Notably, over one thousand differentially expressed genes were identified in HGECs treated with M-PgPs or P. gingivalis with reference to the controls. Gene Ontology and KEGG pathway analysis demonstrated significantly enriched signaling pathways, such as FOXO. Importantly, the FOXO1 inhibitor rescued the M-PgP-induced disruption of cytokine expression. This study suggests that P. gingivalis persisters may perturb innate host defense, through the upregulation of the FOXO signaling pathway. Thus, the current findings could contribute to developing new approaches to tackling P. gingivalis persisters for the effective control of periodontitis and P. gingivalis-related inflammatory comorbidities.


Asunto(s)
Periodontitis , Porphyromonas gingivalis , Citocinas/metabolismo , Células Epiteliales/metabolismo , Humanos , Periodontitis/tratamiento farmacológico , Periodontitis/genética , Periodontitis/metabolismo , Porphyromonas gingivalis/fisiología , Análisis de Secuencia de ARN , Transducción de Señal , Regulación hacia Arriba
18.
Oral Health Prev Dent ; 20(1): 199-206, 2022 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-35481344

RESUMEN

PURPOSE: The role of developmental endothelial locus-1 (DEL-1) in Porphyromonas gingivalis (P. gingivalis)-induced periodontitis and the related molecular mechanisms are unclear. This study aimed to investigate the effect of DEL-1 on SH3 Domain Binding Protein 2 (SH3BP2) expression, and to explore the regulatory role of DEL-1 in periodontal inflammation. MATERIALS AND METHODS: We constructed a P. gingivalis-induced rat experimental periodontitis model, and cultured P. gingivalis-stimulated THP-1 cells in vitro. THP-1 cell viability and cell cycle were examined by CCK-8 and flow cytometry. Rat gingival tissues were collected for hematoxylin-eosin staining. The expression of SH3BP2 and nicotinamide phosphoribosyltransferase (NAMPT) was examined using Western blot. RESULTS: We found that the proliferation of P. gingivalis-infected THP-1 cells was increased by DEL-1. DEL-1 inhibited the expression of NAMPT and SH3BP2 in gingiva tissues of rats with periodontitis as well as in P. gingivalis-infected THP-1 cells. CONCLUSIONS: Overexpression of DEL-1 downregulated SH3BP2 expression and reduced gingival inflammation induced by P. gingivalis. DEL-1 presents some regulatory effects on gingival inflammation in a P. gingivalis-induced rat experimental periodontitis model, suggesting the therapeutic potential of DEL-1 in regulating periodontal inflammation.


Asunto(s)
Gingivitis , Periodontitis , Proteínas Adaptadoras Transductoras de Señales , Animales , Encía , Humanos , Inflamación , Porphyromonas gingivalis/fisiología , Ratas
19.
Periodontol 2000 ; 89(1): 59-82, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35244967

RESUMEN

In the initiation or exacerbation of Alzheimer disease, the dissemination of oral microorganisms into the brain tissue or the low-level systemic inflammation have been speculated to play a role. However, the impact of oral microorganisms, such as Porphyromonas gingivalis, on the pathogenesis of Alzheimer disease and the potential causative relationship is still unclear. The present review has critically reviewed the literature by examining the following aspects: (a) the oral microbiome and the immune response in the elderly population, (b) human studies on the association between periodontal and gut microorganisms and Alzheimer disease, (c) animal and in vitro studies on microorganisms and Alzheimer disease, and (d) preventive and therapeutic approaches. Factors contributing to microbial dysbiosis seem to be aging, local inflammation, systemic diseases, wearing of dentures, living in nursing homes and no access to adequate oral hygiene measures. Porphyromonas gingivalis was detectable in post-mortem brain samples. Microbiome analyses of saliva samples or oral biofilms showed a decreased microbial diversity and a different composition in Alzheimer disease compared to cognitively healthy subjects. Many in-vitro and animal studies underline the potential of P gingivalis to induce Alzheimer disease-related alterations. In animal models, recurring applications of P gingivalis or its components increased pro-inflammatory mediators and ß-amyloid in the brain and deteriorated the animals' cognitive performance. Since periodontitis is the result of a disturbed microbial homoeostasis, an effect of periodontal therapy on the oral microbiome and host response related to cognitive parameters may be suggested and should be elucidated in further clinical trials.


Asunto(s)
Enfermedad de Alzheimer , Microbiota , Anciano , Enfermedad de Alzheimer/etiología , Animales , Disbiosis , Humanos , Inflamación , Microbiota/fisiología , Porphyromonas gingivalis/fisiología
20.
J Immunol Res ; 2022: 6839356, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35224112

RESUMEN

Intestinal bacterial compositions of rheumatoid arthritis (RA) patients have been reported to be different from those of healthy people. Dysbiosis, imbalance of the microbiota, is widely known to cause gut barrier damage, resulting in an influx of bacteria and their substances into host bloodstreams in animal studies. However, few studies have investigated the effect of bacterial substances on the pathophysiology of RA. In this study, eighty-seven active RA patients who had inadequate responses to conventional synthetic disease-modifying antirheumatic drugs or severe comorbidities were analyzed for correlations between many factors such as disease activities, disease biomarkers, intestinal bacterial counts, fecal and serum lipopolysaccharide (LPS), LPS-binding protein (LBP), endotoxin neutralizing capacity (ENC), and serum antibacterial substance IgG and IgA antibody levels by multiple regression analysis with consideration for demographic factors such as age, sex, smoking, and methotrexate treatment. Serum LBP levels, fecal LPS levels, total bacteria counts, serum anti-LPS from Porphyromonas gingivalis (Pg-LPS) IgG antibody levels, and serum anti-Pg-LPS IgA antibody levels were selected for multiple regression analysis using Spearman's correlation analysis. Serum LBP levels were correlated with disease biomarker levels, such as erythrocyte sedimentation rate (p < 0.001), C-reactive protein (p < 0.001), matrix metalloproteinase-3 (p < 0.001), and IL-6 (p = 0.001), and were inversely correlated with hemoglobin (p = 0.005). Anti-Pg-LPS IgG antibody levels were inversely correlated with activity indices such as patient global assessments using visual analogue scale (VAS) (p = 0.002) and painVAS (p < 0.001). Total bacteria counts were correlated with ENC (p < 0.001), and inversely correlated with serum LPS (p < 0.001) and anti-Pg-LPS IgA antibody levels (p < 0.001). These results suggest that substances from oral and gut microbiota may influence disease activity in RA patients.


Asunto(s)
Artritis Reumatoide/microbiología , Infecciones por Bacteroidaceae/microbiología , Disbiosis/microbiología , Boca/microbiología , Porphyromonas gingivalis/fisiología , Proteínas de Fase Aguda/metabolismo , Anciano , Artritis Reumatoide/inmunología , Autoanticuerpos/sangre , Carga Bacteriana , Infecciones por Bacteroidaceae/inmunología , Biomarcadores/metabolismo , Proteínas Portadoras/metabolismo , Estudios Transversales , Disbiosis/inmunología , Femenino , Microbioma Gastrointestinal , Humanos , Inmunoglobulina A/metabolismo , Lipopolisacáridos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...