Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Immunol Res ; 11(3): 290-305, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36633576

RESUMO

Accumulating evidence shows that PD-L1 expression on dendritic cells (DC) is critical for cancer immunotherapy and that Porphyromonas gingivalis (Pg) colonization aggravates the progression of upper gastrointestinal cancers. However, the effects of Pg infection on PD-L1 expression on DCs and related immune consequences in the infection milieu of oral cancer remain unexplored. Here, we found that Pg infection robustly enhanced PD-L1 expression on DCs in a gingipain-dependent manner in cultured cell and systemic infection assays. Pg infection suppressed antigen-specific CD8+ T cells through upregulation of PD-L1 expression on ovalbumin (OVA)-pulsed DCs. This suppression was manifested by decreased IFNγ, perforin, granzyme B, and CD107a. Further analysis showed that Pg drastically reduced CD8+ T cells' ability to lyse OVA-pulsed target cells. Additionally, Pg infection increased the phosphorylation of Akt and STAT3, leading to a significant increase in PD-L1 expression. This was substantiated by using siRNA, overexpression plasmids, and pharmacologic inhibitors. Consistent with the in vitro observations, in a syngeneic mouse oral cancer model, Pg infection significantly enhanced PD-L1 expression on DCs from intratumoral tissues and cervical lymph nodes and exacerbated oral cancer progression, whereas a Pg lysine-specific, gingipain-defective mutant failed to do so. These influences of Pg were largely diminished when tumor cells were pretreated with antibiotics or a STAT3 inhibitor. Therefore, we demonstrated that Pg infection upregulates PD-L1 expression on DCs through Akt-STAT3 signaling, suppresses CD8+ T-cell cytotoxicity, and aggravates oral cancer growth, suggesting targeting Pg, and/or its mediated signaling, could be a therapeutic strategy to improve the efficacy of checkpoint blockade immunotherapy.


Assuntos
Antígeno B7-H1 , Neoplasias Bucais , Animais , Camundongos , Cisteína Endopeptidases Gingipaínas/metabolismo , Cisteína Endopeptidases Gingipaínas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linfócitos T CD8-Positivos , Células Dendríticas
2.
FEBS Lett ; 595(11): 1604-1612, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33792027

RESUMO

Streptococcus pneumoniae causes pneumonia by infecting the alveolar epithelium via binding to host receptors, such as the platelet-activating factor receptor (PAFR). Although chronic periodontitis has been identified as a pneumonia risk factor, how periodontopathic bacteria cause pneumonia is not known. We found that S. pneumoniae adhered to PAFR expressed on A549 human alveolar epithelial cells stimulated by Porphyromonas gingivalis culture supernatant, and this was abrogated by a PAFR-specific inhibitor. Among the major virulence factors of P. gingivalis [lipopolysaccharide (LPS), fimbriae and gingipains (Rgps and Kgp)], PAFR expression and pneumococcal adhesion were executed in an Rgp-dependent manner. LPS and fimbriae did not induce PAFR expression. Hence, our findings suggest that P. gingivalis enhances pneumococcal adhesion to human alveoli by inducing PAFR expression and that gingipains are responsible for this.


Assuntos
Cisteína Endopeptidases Gingipaínas/farmacologia , Glicoproteínas da Membrana de Plaquetas/genética , Porphyromonas gingivalis/metabolismo , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G/genética , Fatores de Virulência/farmacologia , Células A549 , Aderência Bacteriana/efeitos dos fármacos , Técnicas de Cocultura , Meios de Cultivo Condicionados/química , Meios de Cultivo Condicionados/farmacologia , Fímbrias Bacterianas/química , Regulação da Expressão Gênica , Cisteína Endopeptidases Gingipaínas/deficiência , Cisteína Endopeptidases Gingipaínas/genética , Interações Hospedeiro-Patógeno/genética , Humanos , Lipopolissacarídeos/farmacologia , Modelos Biológicos , Glicoproteínas da Membrana de Plaquetas/agonistas , Glicoproteínas da Membrana de Plaquetas/metabolismo , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/patogenicidade , Alvéolos Pulmonares/microbiologia , RNA Mensageiro/agonistas , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Virulência/deficiência , Fatores de Virulência/genética
3.
Sci Rep ; 10(1): 21109, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33273542

RESUMO

Porphyromonas gingivalis, a periodontal pathogen, translocates many virulence factors including the cysteine proteases referred to as gingipains to the cell surface via the type IX secretion system (T9SS). Expression of the T9SS component proteins is regulated by the tandem signaling of the PorXY two-component system and the ECF sigma factor SigP. However, the details of this regulatory pathway are still unknown. We found that one of the T9SS conserved C-terminal domain-containing proteins, PGN_0123, which we have designated PorA, is involved in regulating expression of genes encoding T9SS structural proteins and that PorA can be translocated onto the cell surface without the T9SS translocation machinery. X-ray crystallography revealed that PorA has a domain similar to the mannose-binding domain of Escherichia coli FimH, the tip protein of Type 1 pilus. Mutations in the cytoplasmic domain of the sensor kinase PorY conferred phenotypic recovery on the ΔporA mutant. The SigP sigma factor, which is activated by the PorXY two-component system, markedly decreased in the ΔporA mutant. These results strongly support a potential role for PorA in relaying a signal from the cell surface to the PorXY-SigP signaling pathway.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos , Porphyromonas gingivalis/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Mutação com Ganho de Função , Cisteína Endopeptidases Gingipaínas/farmacologia , Hemaglutinação/efeitos dos fármacos , Lipopolissacarídeos/metabolismo , Modelos Biológicos , Mapeamento de Peptídeos , Pigmentação/efeitos dos fármacos , Porphyromonas gingivalis/efeitos dos fármacos , Domínios Proteicos , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
4.
Front Immunol ; 11: 1551, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32793214

RESUMO

Background:Porphyromonas gingivalis and its inflammagens are associated with a number of systemic diseases, such as cardiovascular disease and type 2 diabetes (T2DM). The proteases, gingipains, have also recently been identified in the brains of Alzheimer's disease patients and in the blood of Parkinson's disease patients. Bacterial inflammagens, including lipopolysaccharides (LPSs) and various proteases in circulation, may drive systemic inflammation. Methods: Here, we investigate the effects of the bacterial products LPS from Escherichia coli and Porphyromonas gingivalis, and also the P. gingivalis gingipain [recombinant P. gingivalis gingipain R1 (RgpA)], on clot architecture and clot formation in whole blood and plasma from healthy individuals, as well as in purified fibrinogen models. Structural analysis of clots was performed using confocal microscopy, scanning electron microscopy, and AFM-Raman imaging. We use thromboelastography® (TEG®) and rheometry to compare the static and dynamic mechanical properties of clots. Results: We found that these inflammagens may interact with fibrin(ogen) and this interaction causes anomalous blood clotting. Conclusions: These techniques, in combination, provide insight into the effects of these bacterial products on cardiovascular health, and particularly clot structure and mechanics.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Cisteína Endopeptidases Gingipaínas/química , Cisteína Endopeptidases Gingipaínas/farmacologia , Fenômenos Mecânicos , Porphyromonas gingivalis/enzimologia , Adulto , Feminino , Fibrina/química , Fibrinogênio/química , Fibrinogênio/ultraestrutura , Humanos , Lipopolissacarídeos/efeitos adversos , Masculino , Microscopia de Força Atômica , Microscopia Confocal , Pessoa de Meia-Idade , Proteínas Recombinantes , Reologia , Análise Espectral Raman , Trombose/tratamento farmacológico , Adulto Jovem
5.
J Mol Histol ; 51(2): 147-159, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32193744

RESUMO

As a crucial virulence factor of Porphyromonas gingivalis, gingipains play an important role in periodontal destruction. This study aimed to investigate the effect of gingipains on osteoclastogenesis. We used RAW264.7 cells as osteoclast precursors in our study. In experimental groups, cells were treated with gingipains and/or receptor activator of nuclear factor-κB ligand (RANKL). Tartrate-resistant acid phosphatase (TRAP) activity staining assay showed osteoclast precursors and RANKL-induced mature osteoclasts were increased in a gingipains dose-dependent manner. Real-time reverse transcription polymerase chain reaction analysis demonstrated that gingipains upregulated osteoclastic genes including the protease cathepsin K (Ctsk), matrix metalloprotein 9 (Mmp9), nuclear factor of activated T cells 1 (Nfatc1) and acid phosphatase 5, tartrate resistant (Acp5) in a time-dependent manner. Western blotting assays presented upregulated expressions of TNF receptor-activating factor 6 (TRAF6) and integrin ß3 induced by gingipains and RANKL compared to RANKL alone. Enhanced integrin-related signaling was also demonstrated by elevated phosphorylations of FAK and paxillin compared to control. Moreover, the pit resorption assays showed that gingipains augmented bone resorptive function of osteoclasts induced by RANKL. When we used Cilengitide to block integrin αvß3, gingipains reversed the reduction of formation and resorptive function in RANKL-induced osteoclasts, as they enhanced integrin αvß3 levels more than RANKL treatment alone. In conclusion, our data suggest that gingipains augmented the differentiation and function of mature osteoclasts induced by RANKL through the increase in integrin αvß3.


Assuntos
Cisteína Endopeptidases Gingipaínas/farmacologia , Integrina beta3/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Ligante RANK/farmacologia , Animais , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Integrina beta3/genética , Camundongos , Osteoclastos/citologia , Paxilina/metabolismo , Fosforilação , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , Fator 6 Associado a Receptor de TNF/metabolismo , Fosfatase Ácida Resistente a Tartarato/metabolismo
6.
Eur J Oral Sci ; 127(4): 287-293, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31175838

RESUMO

Arginine-specific cysteine proteinases, such as Arg-gingipain B (RgpB), mediate inflammation by activating protease-activated receptors (PARs). Arg-gingipain B is produced by Porphyromonas gingivalis, and is implicated in the causation of periodontal disease. The purpose of the present study was to observe the influence of recombinant RgpB protein (rRgpB) on PAR activation by monitoring intracellular Ca2+ ion concentration ([Ca2+]i) and inositol-1,4,5-triphosphate (IP3) levels in human gingival fibroblasts (HGFs). Our findings showed that rRgpB could cause a transient increase in [Ca2+]i. This increase in [Ca2+]i was completely suppressed by vorapaxar, a PAR-1 antagonist. Recombinant Arg-gingipain B increased the concentration of IP3, reaching a maximum at 60 s after treatment; this was completely inhibited by vorapaxar. We therefore conclude that rRgpB-induced calcium signaling in HGFs is mainly caused by PAR-1 activation. This suggests that PAR-1 activation plays a significant role in chronic inflammatory periodontal disease induced by P. gingivalis RgpB.


Assuntos
Sinalização do Cálcio , Fibroblastos/metabolismo , Cisteína Endopeptidases Gingipaínas/farmacologia , Porphyromonas gingivalis/enzimologia , Receptor PAR-1/metabolismo , Proteínas de Bactérias/farmacologia , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Humanos , Inositol 1,4,5-Trifosfato , Lactonas/farmacologia , Piridinas/farmacologia , Receptor PAR-1/antagonistas & inibidores , Proteínas Recombinantes/farmacologia
7.
Sci Adv ; 5(1): eaau3333, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30746447

RESUMO

Porphyromonas gingivalis, the keystone pathogen in chronic periodontitis, was identified in the brain of Alzheimer's disease patients. Toxic proteases from the bacterium called gingipains were also identified in the brain of Alzheimer's patients, and levels correlated with tau and ubiquitin pathology. Oral P. gingivalis infection in mice resulted in brain colonization and increased production of Aß1-42, a component of amyloid plaques. Further, gingipains were neurotoxic in vivo and in vitro, exerting detrimental effects on tau, a protein needed for normal neuronal function. To block this neurotoxicity, we designed and synthesized small-molecule inhibitors targeting gingipains. Gingipain inhibition reduced the bacterial load of an established P. gingivalis brain infection, blocked Aß1-42 production, reduced neuroinflammation, and rescued neurons in the hippocampus. These data suggest that gingipain inhibitors could be valuable for treating P. gingivalis brain colonization and neurodegeneration in Alzheimer's disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/microbiologia , Infecções por Bacteroidaceae/tratamento farmacológico , Encéfalo/microbiologia , Encéfalo/patologia , Fármacos Neuroprotetores/uso terapêutico , Porphyromonas gingivalis/enzimologia , Bibliotecas de Moléculas Pequenas/uso terapêutico , Idoso , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Infecções por Bacteroidaceae/microbiologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Cisteína Endopeptidases Gingipaínas/antagonistas & inibidores , Cisteína Endopeptidases Gingipaínas/metabolismo , Cisteína Endopeptidases Gingipaínas/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/metabolismo , Projetos Piloto , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/genética , Estudos Prospectivos , Saliva/microbiologia , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA