Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Innate Immun ; 14(4): 306-319, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34823251

RESUMO

Plasminogen activator inhibitor-1 (PAI-1), a serine protease inhibitor, is constitutively produced by endothelial cells and plays a vital role in maintaining vascular homeostasis. Chronic periodontitis is an inflammatory disease characterized by bleeding of periodontal tissues that support the tooth. In this study, we aimed to determine the role of PAI-1 produced by endothelial cells in response to infections caused by the primary periodontal pathogen Porphyromonas gingivalis. We demonstrated that P. gingivalis infection resulted in significantly reduced PAI-1 levels in human endothelial cells. This reduction in PAI-1 levels could be attributed to the proteolysis of PAI-1 by P. gingivalis proteinases, especially lysine-specific gingipain-K (Kgp). We demonstrated the roles of these degradative enzymes in the endothelial cells using a Kgp-specific inhibitor and P. gingivalis gingipain-null mutants, in which the lack of the proteinases resulted in the absence of PAI-1 degradation. The degradation of PAI-1 by P. gingivalis induced a delayed wound healing response in endothelial cell layers via the low-density lipoprotein receptor-related protein. Our results collectively suggested that the proteolysis of PAI-1 in endothelial cells by gingipains of P. gingivalis might lead to the deregulation of endothelial homeostasis, thereby contributing to the permeabilization and dysfunction of the vascular endothelial barrier.


Assuntos
Células Endoteliais , Porphyromonas gingivalis , Adesinas Bacterianas/metabolismo , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/farmacologia , Cisteína Endopeptidases Gingipaínas , Humanos , Inibidor 1 de Ativador de Plasminogênio , Porphyromonas gingivalis/fisiologia , Cicatrização
2.
J Periodontol ; 91(1): 129-137, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31347700

RESUMO

BACKGROUND: Serum amyloid A (SAA) has been identified to trigger inflammation response, and play a crucial role in chronic inflammatory diseases. However, the regulatory mechanism of SAA still remains unclear during the development of periodontitis METHODS: SAA mRNA and protein expression were detected in healthy and inflammatory gingival tissues using real-time polymerase chain reaction (PCR) and immunohistochemistry. Human recombinant SAA (Apo-SAA), Pam3CSK4 (a Toll-like receptor (TLR) 2 ligand), siRNA-SAA, or TLR2 neutralizing antibody was applied to treat human gingival fibroblasts, respectively, or combined. SAA, TLRs, and inflammatory cytokines interleukin (IL)-6 and IL-8 were analyzed by real-time PCR, western blotting, or enzyme-linked immunosorbent assay. RESULTS: SAA expression increased in human inflammatory gingival tissues from patients with periodontitis (P <0.05). Apo-SAA could increase not only the mRNA expression of TLR2 (P <0.05), but also IL-6 and IL-8 mRNA and protein levels (P <0.05) which was suppressed by TLR2 antibody in human gingival fibroblasts. Pam3CSK4 increased SAA, IL-6, and IL-8 levels (P <0.05). However, the expression of SAA, IL-6, and IL-8 decreased after transfection of siRNA-SAA (P <0.05). CONCLUSION: SAA not only increases in inflammatory gingiva, but also triggers inflammatory cytokine secretion via interacting with TLR2 pathway in human gingival fibroblasts, which indicates that SAA is involved in periodontal inflammation.


Assuntos
Gengiva , Receptor 2 Toll-Like , Células Cultivadas , Citocinas , Fibroblastos , Humanos , Proteína Amiloide A Sérica
3.
J Phys Chem B ; 123(35): 7570-7577, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31401833

RESUMO

The investigation on proteinlike specific functions of nanoparticles (NPs) has been a huge challenge. Here, the biocompatibility of Au nanoparticles (AuNPs) to antigens hen egg white lysozyme and epidermal growth factor receptor was studied first by molecular dynamics (MD) simulations and the research results revealed that antigens could form quickly a stable binding with the AuNPs and kept the structural integrity of the protein, which demonstrated better biocompatibility of AuNPs. Then, two types of complementary-determining regions (CDRs) were grafted onto the AuNPs to design a novel multi-CDR-functional nanobody. By means of MD simulations under physiological conditions, we found that the bindings of the designed nanobody and the antigens were stable and safe. Compared with the results of antigens interacting with the natural antibody, the redundant CDRs on AuNPs bound with the nonactive site in the antigens to form a stable conformation, which leaded to the powerful binding capacity of the designed nanobody than that of the natural antibody. This study provided available insights into the biocompatibility of AuNPs and important theoretical proofs to the multi-CDR-functional nanobody applied in biological systems, which were expected to help in design of novel multifunctional nanobodies.


Assuntos
Materiais Biocompatíveis/química , Ouro/química , Nanopartículas Metálicas/química , Simulação de Dinâmica Molecular , Muramidase/química , Receptores ErbB/química , Humanos , Muramidase/metabolismo
4.
J Periodontol ; 88(11): e179-e187, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28598282

RESUMO

BACKGROUND: MicroRNAs (miRs) play a crucial role in inflammatory diseases, including periodontitis. Meanwhile, miRs act as biomarkers for predicting diabetes mellitus (DM). However, the regulatory mechanism of miR-126 on development of periodontitis in patients with DM still remains unclear. METHODS: Human gingival fibroblasts were cultured with low (5.5 mmol/L), medium (15 mmol/L), and high (25 mmol/L) glucose, respectively. Expressions of miR-126, tumor necrosis factor (TNF) receptor associated factor (TRAF) 6, and related cytokines were analyzed by real-time polymerase chain reaction (PCR). After transfection with miR-126 mimic, PCR and western blot were performed to detect level of TRAF6, and luciferase reporter assay confirmed if TRAF6 is the direct target of miR-126. Production of cytokines was measured using enzyme-linked immunosorbent assay. RESULTS: Increased glucose significantly suppressed miR-126 expression in human gingival fibroblasts (P <0.05). Also, high glucose increased TRAF6, interleukin (IL)-6, TNF-α, and chemical chemokine ligand (CCL) 2 levels, whereas it decreased IL-10 level. MiR-126 mimic significantly decreased TRAF6 mRNA and protein levels under high glucose (P <0.05). Also, miR-126 directly targeted TRAF6 through binding to its 3' untranslated region in human gingival fibroblasts. Overexpression of miR-126 significantly abrogated high glucose-induced secretion of proinflammatory cytokines such as IL-6, TNF-α, and CCL2 and promoted production of IL-10. CONCLUSION: These data suggest that miR-126 inhibits inflammation of human gingival fibroblasts under high glucose through targeting TRAF6, which may be a potential therapeutic target for periodontitis concomitant with DM.


Assuntos
Citocinas/metabolismo , Fibroblastos/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Glucose/farmacologia , MicroRNAs/fisiologia , Fator 6 Associado a Receptor de TNF/fisiologia , Adolescente , Adulto , Western Blotting , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Fibroblastos/metabolismo , Gengiva/metabolismo , Humanos , Masculino , Reação em Cadeia da Polimerase em Tempo Real , Fator 6 Associado a Receptor de TNF/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA