Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Oral Dis ; 29(8): 3571-3582, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35765229

RESUMO

OBJECTIVES: Ferroptosis is associated with multiple inflammatory diseases. Periodontitis is an inflammatory disease mainly caused by oral opportunistic pathogens. However, the ferroptosis-periodontitis relationship has not been thoroughly described. We here analyzed whether ferroptosis is involved in periodontitis. MATERIALS AND METHODS: Human gingival fibroblasts (HGFs) were stimulated with P. gingivalis-LPS and ferrostatin-1 (Fer-1, a ferroptosis inhibitor), and changes in mitochondrial morphology, ferroptosis-related factors, and inflammation levels were detected. After the rat experimental periodontitis model was established, changes in ferroptosis-related factors and inflammation levels were re-evaluated in the same manner. RESULTS: Porphyromonas gingivalis-LPS-induced mitochondrial shrinkage, an increase in mitochondrial membrane density, and upregulation of reactive oxygen species in HGFs. The expression of prostaglandin-endoperoxide synthase 2, transferrin receptor 1, and malondialdehyde and inflammation levels were upregulated, whereas the expression of solute carrier family seven member 11, glutathione peroxidase 4, superoxide dismutase, and glutathione were downregulated. Fer-1 attenuated these aforementioned changes and inflammation levels induced by P. gingivalis-LPS. The in vivo experiment results were consistent with the in vitro experiment results. CONCLUSIONS: Ferroptosis is involved in inflammatory processes in HGFs upon P. gingivalis-LPS stimulation. Ferroptosis is observed in the gingival tissue of periodontitis rats.


Assuntos
Ferroptose , Periodontite , Humanos , Animais , Ratos , Lipopolissacarídeos/farmacologia , Porphyromonas gingivalis/metabolismo , Periodontite/metabolismo , Inflamação/metabolismo , Gengiva/metabolismo , Fibroblastos , Células Cultivadas
2.
Mol Biol Rep ; 49(7): 5821-5829, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35716284

RESUMO

BACKGROUND: Peri-implantitis is the main cause of dental implant failure, which is associated with pyroptosis. The roles of D-aspartic acid (D-Asp) on pyroptosis and the mechanism of the protective effect of D-Asp on human gingival fibroblasts (HGFs) remain unknown. This study investigated the effects of D-Asp on the pyroptosis of HGFs induced by high mobility group box 1 protein (HMGB1). METHODS: The cytotoxic effects of D-Asp on HGFs was detected by Cell Counting Kit-8 assay, the membrane permeability was investigated by propidium iodide/ Hoechst 33,342 double staining, flow cytometry analysis, and lactate dehydrogenase releasing, The gene and protein expression levels were detected by real-time quantitative PCR, enzyme-linked immunosorbent assay, and Western blot, respectively. RESULTS: Cell viability analysis showed that D-Asp ≤ 30 mM had no cytotoxicity to HGFs. HMGB1 drastically raised the membrane permeability of HGFs, while 1/10/30 mM D-Asp suppressed the permeability and remained the integrity of the membrane. HMGB1 promoted the mRNA expression of NLRP3, caspase-1, GSDMD, IL-1ß, and IL-18, and the protein expression of IL-1ß, IL-18, caspase-1, GSDMD, and NLRP3. CONCLUSIONS: With the pretreatment of HGFs with D-Asp of 1/10/30 mM for 24 h, the cell membrane permeability was reduced and the expression of NLRP3, caspase-1, GSDMD, IL-1ß, and IL-18 was significantly decreased compared with the HMGB1 group, indicating the competitive antagonism of D-Asp against HMGB1 on the binding with toll-like receptors. Hence, this study may provide a novel insight into preventing pyroptosis and propose a new strategy for the treatment of peri-implantitis.


Assuntos
Proteína HMGB1 , Peri-Implantite , Caspase 1/metabolismo , Ácido D-Aspártico/farmacologia , Fibroblastos/metabolismo , Proteína HMGB1/metabolismo , Humanos , Inflamação , Interleucina-18 , Interleucina-1beta/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose
3.
Biofouling ; 37(2): 222-234, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33682548

RESUMO

Porphyromonas gingivalis biofilms are implicated in the pathology of peri-implantitis and periodontitis. In this study, D-arginine (R), D-methionine (M), D-histidine (H), and a mixture of these D-amino acids (D-AAs) were investigated as an effective therapeutic strategy against P. gingivalis biofilms. The bacterial growth activity and minimum inhibitory concentrations were determined for each D-AA, along with the effects of the D-AAs mixture on biofilm development, morphology, structure, extracellular polysaccharides (EPS), cytotoxicity towards commensals, and bacterial structure. The D-AA mixture delayed the proliferation of P. gingivalis, changed its membrane structure, and decreased biofilm thickness and integrity, as compared with individual D-AAs. The EPS content increased with the concentration of D-AAs. The present study shows that a 4 mM RMH, triple D-AA mixture, enhanced deleterious effects on P. gingivalis biofilms without any cytotoxicity compared with individual D-AAs, thus providing a new strategy for the treatment of peri-implantitis and periodontitis.


Assuntos
Histidina , Porphyromonas gingivalis , Arginina , Biofilmes , Metionina
4.
Front Bioeng Biotechnol ; 10: 864012, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35309977

RESUMO

Peri-implantitis is the leading cause of dental implant failure, initially raised by biofilm accumulation on the implant surface. During the development of biofilm, Actinomyces viscosus (A. viscosus) plays a pivotal role in initial attachment as well as the bacterial coaggregation of multispecies pathogens. Hence, eliminating the A. viscosus-associated biofilm is fundamental for the regeneration of the lost bone around implants. Whereas clinical evidence indicated that antimicrobials and debridement did not show significant effects on the decontamination of biofilm on the implant surface. In this study, alpha-amylase was investigated for its effects on disassembling A. viscosus biofilm. Then, in order to substantially disperse biofilm under biosafety concentration, D-arginine was employed to appraise its enhancing effects on alpha-amylase. In addition, molecular dynamics simulations and molecular docking were conducted to elucidate the mechanism of D-arginine enhancing alpha-amylase. 0.1-0.5% alpha-amylase showed significant effects on disassembling A. viscosus biofilm, with definite cytotoxicity toward MC3T3-E1 cells meanwhile. Intriguingly, 8 mM D-arginine drastically enhanced the eradication of A. viscosus biofilm biomass by 0.01% alpha-amylase with biosafety in 30 min. The exopolysaccharides of biofilm were also thoroughly hydrolyzed by 0.01% alpha-amylase with 8 mM D-arginine. The biofilm thickness and integrity were disrupted, and the exopolysaccharides among the extracellular matrix were elusive. Molecular dynamics simulations showed that with the hydrogen bonding of D-arginine to the catalytic triad and calcium-binding regions of alpha-amylase, the atom fluctuation of the structure was attenuated. The distances between catalytic triad were shortened, and the calcium-binding regions became more stable. Molecular docking scores revealed that D-arginine facilitated the maltotetraose binding process of alpha-amylase. In conclusion, these results demonstrate that D-arginine enhances the disassembly effects of alpha-amylase on A. viscosus biofilm through potentiating the catalytic triad and stabilizing the calcium-binding regions, thus providing a novel strategy for the decontamination of biofilm contaminated implant surface.

5.
ACS Appl Mater Interfaces ; 14(48): 53523-53534, 2022 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-36401828

RESUMO

Autologous blood-derived protein hydrogels have shown great promise in the field of personalized regenerative medicine. However, the inhospitable regenerative microenvironments, especially the unfavorable immune microenvironment, are closely associated with their limited tissue-healing outcomes. Herein, novel immunomodulatory blood-derived hybrid hydrogels (PnP-iPRF) are rationally designed and constructed for enhanced bone regeneration via multichannel regulation of the osteogenic microenvironment. Such double-network hybrid hydrogels are composed of clinically approved injectable platelet-rich fibrin (i-PRF) and polycaprolactone/hydroxyapatite composite nanofibers by using enriched polydopamine (PDA) as the anchor. The polycaprolactone component in PnP-iPRF provides a reinforced structure to stimulate osteoblast differentiation in a proper biomechanical microenvironment. Most importantly, the versatile PDA component in PnP-iPRF can not only offer high adhesion capacity to the growth factors of i-PRF and create a suitable biochemical microenvironment for sustained osteogenesis but also reprogram the osteoimmune microenvironment via the induction of M2 macrophage polarization to promote bone healing. The present study will provide a new paradigm to realize enhanced osteogenic efficacy by multichannel microenvironment regulations and give new insights into engineering high-efficacy i-PRF hydrogels for regenerative medicine.


Assuntos
Regeneração Óssea , Hidrogéis , Hidrogéis/farmacologia
6.
Inflammation ; 44(3): 846-858, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33140204

RESUMO

Periodontitis is a chronic inflammatory disease induced by Porphyromonas gingivalis (P. gingivalis) and other pathogens. P. gingivalis release various virulence factors including lipopolysaccharide (LPS). However, whether P. gingivalis-LPS inducing pyroptosis in human gingival fibroblasts (HGFs) remains unknown. In present study, P. gingivalis-LPS decreased the membrane integrity of HGFs, and pyroptosis-associated cytokines were upregulated at the mRNA level. In addition, pyroptosis proteins were highly expressed in gingival tissues of periodontitis. P. gingivalis-LPS induced gingivitis in the rat model, and the expression level of pyroptosis-associated proteins increased. Together, P. gingivalis-LPS can activate the pyroptosis reaction, which may be a pro-pyroptosis status in a relative low concentration.


Assuntos
Fibroblastos/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Gengivite/induzido quimicamente , Lipopolissacarídeos/toxicidade , Porphyromonas gingivalis/metabolismo , Piroptose/efeitos dos fármacos , Animais , Caspase 1/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Gengiva/metabolismo , Gengiva/patologia , Gengivite/metabolismo , Gengivite/patologia , Humanos , Lipopolissacarídeos/isolamento & purificação , Masculino , Ratos Sprague-Dawley , Transdução de Sinais , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA