Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int Dent J ; 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38685137

RESUMO

PURPOSE: To explore the mechanism underlying autophagy disruption in gingival epithelial cells (GECs) in diabetic individuals. METHODS AND MATERIALS: Bone marrow-derived macrophages (BMDMs) and GECs were extracted from C57/bl and db/db mice, the exosomes (Exo) were isolated from BMDMs. qRT‒PCR and Western blotting were performed to analyse gene expression. The AnimalTFDB database was used to identify relevant transcription factors, and miRNA sequencing was utilised to identify relevant miRNAs with the aid of the TargetScan/miRDB/miRWalk databases. A dual-luciferase assay was conducted to verify intermolecular targeting relationships. RESULTS: Similar to BMDMs, BMDM-derived Exos disrupted autophagy and exerted proinflammatory effects in GEC cocultures, and ATG7 may play a vital role. AnimalTFDB database analysis and dual-luciferase assays indicated that NR5A2 is the most relevant transcription factor that regulates Atg7 expression. SiRNA-NR5A2 transfection blocked autophagy in GECs and exacerbated inflammation, whereas NR5A2 upregulation restored ATG7 expression and ameliorated ExoDM-mediated inflammation. MiRNA sequencing, with TargetScan/miRDB/miRWalk analyses and dual-luciferase assays, confirmed that miR-381-3p is the most relevant miRNA that targets NR5A2. MiR-381-3p mimic transfection blocked autophagy in GECs and exacerbated inflammation, while miR-381-3p inhibitor transfection restored ATG7 expression and attenuated ExoDM-mediated inflammation. CONCLUSION: BMDM-derived Exos, which carry miR-381-3p, inhibit NR5A2 and disrupt autophagy in GECs, increasing periodontal inflammation in diabetes.

2.
Int Immunopharmacol ; 128: 111465, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38181674

RESUMO

Periodontitis (PD) is a common chronic oral inflammatory disease that cause alveolar bone loss. Current strategies for bone regeneration achieve limited results in PD. The aberrant host osteoimmunity to pathogenic bacteria is responsible for the destruction of alveolar bone in PD. We aimed to investigate the distinctive activity of immune cells in PD to create more effective and precise therapeutic approaches for treating PD. In this study, we revealed that neutrophils in the inflamed alveolar bone of PD patients expressed higher levels of CXCR1/2 and had a stronger pro-inflammatory capacity and chemotactic ability than that in healthy individuals. Suppressing the recruitment of neutrophils to inflamed sites with the CXCR1/2 inhibitor reparixin reduced alveolar bone loss in PD mice. In this study, we not only revealed that neutrophils exhibit a heterogeneously stronger pro-inflammatory capacity in the inflamed alveolar bone of PD patients but also provided a precise therapeutic treatment for PD involving the suppression of neutrophil recruitment.


Assuntos
Perda do Osso Alveolar , Periodontite , Humanos , Camundongos , Animais , Perda do Osso Alveolar/patologia , Infiltração de Neutrófilos , Neutrófilos , Bactérias
3.
Innovation (Camb) ; 5(1): 100547, 2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38170012

RESUMO

Cognitive decline has been linked to periodontitis through an undetermined pathophysiological mechanism. This study aimed to explore the mechanism underlying periodontitis-related cognitive decline and identify therapeutic strategies for this condition. Using single-nucleus RNA sequencing we found that changes in astrocyte number, gene expression, and cell‒cell communication were associated with cognitive decline in mice with periodontitis. In addition, activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome was observed to decrease the phagocytic capability of macrophages and reprogram macrophages to a more proinflammatory state in the gingiva, thus aggravating periodontitis. To further investigate this finding, lipid-based nanoparticles carrying NLRP3 siRNA (NPsiNLRP3) were used to inhibit overactivation of the NLRP3 inflammasome in gingival macrophages, restoring the oral microbiome and reducing periodontal inflammation. Furthermore, gingival injection of NPsiNLRP3 reduced the number of Serpina3nhigh astrocytes in the hippocampus and prevented cognitive decline. This study provides a functional basis for the mechanism by which the destruction of periodontal tissues can worsen cognitive decline and identifies nanoparticle-mediated restoration of gingival macrophage function as a novel treatment for periodontitis-related cognitive decline.

4.
Biomater Sci ; 11(9): 3214-3226, 2023 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-36927992

RESUMO

The severe pain caused by oral ulcers seriously affects food intake and speech, bringing great inconvenience in daily life. Drug-loaded patches are mostly used to treat oral mucosal diseases such as oral ulcers and oral lichen planus, but their effects are limited because of the influences of saliva and muscle movement. To enhance the adhesion of drug-loaded patches used in the oral cavity, we designed antimicrobial peptides (AMPs)-modified polycaprolactone (PCL)-collagen nanosheets (APCNs). The internal layer is a bioactive and antibacterial collagen layer modified with antimicrobial peptides. The backing layer is a hydrophobic PCL layer with good mechanical strength that can reduce external influences. We have characterized and tested the APCNs. First, the APCNs exhibited continuous and strong adhesion to irregular buccal mucosa surfaces under wet conditions and external force action. Antibacterial experiments showed that the APCNs had high antibacterial activity against both Gram-positive bacteria and Gram-negative bacteria. Moreover, the APCNs showed good biocompatibility and promoted the adhesion of fibroblasts in vitro. Furthermore, APCNs treatment accelerated ulcer healing in a Sprague Dawley rat oral ulcer model. Our study developed antibacterial, wet-adhesive, and healing-promoting PCL-collagen nanosheets and demonstrated that these nanosheets could be promising adhesive therapeutic agents for the treatment of oral mucosal ulcers.


Assuntos
Úlceras Orais , Ratos , Animais , Úlceras Orais/tratamento farmacológico , Adesivos/farmacologia , Ratos Sprague-Dawley , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Antibacterianos/química , Úlcera/tratamento farmacológico , Colágeno/uso terapêutico
5.
Int J Oral Sci ; 13(1): 43, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34907166

RESUMO

Although mesenchymal stem cell-derived exosomes (MSC-exos) have been shown to have therapeutic effects in experimental periodontitis, their drawbacks, such as low yield and limited efficacy, have hampered their clinical application. These drawbacks can be largely reduced by replacing the traditional 2D culture system with a 3D system. However, the potential function of MSC-exos produced by 3D culture (3D-exos) in periodontitis remains elusive. This study showed that compared with MSC-exos generated via 2D culture (2D-exos), 3D-exos showed enhanced anti-inflammatory effects in a ligature-induced model of periodontitis by restoring the reactive T helper 17 (Th17) cell/Treg balance in inflamed periodontal tissues. Mechanistically, 3D-exos exhibited greater enrichment of miR-1246, which can suppress the expression of Nfat5, a key factor that mediates Th17 cell polarization in a sequence-dependent manner. Furthermore, we found that recovery of the Th17 cell/Treg balance in the inflamed periodontium by the local injection of 3D-exos attenuated experimental colitis. Our study not only showed that by restoring the Th17 cell/Treg balance through the miR-1246/Nfat5 axis, the 3D culture system improved the function of MSC-exos in the treatment of periodontitis, but also it provided a basis for treating inflammatory bowel disease (IBD) by restoring immune responses in the inflamed periodontium.


Assuntos
Colite , Exossomos , Periodontite , Humanos , Periodontite/terapia , Periodonto , Linfócitos T Reguladores , Células Th17
6.
Biomaterials ; 275: 120963, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34153785

RESUMO

Impaired wound healing presents great health risks to patients. While encouraging, the current clinical successes of mesenchymal stromal cell (MSC)-based therapies for tissue repair have been limited. Genetic engineering could endow MSCs with more robust regenerative capacities. Here, we identified that C-C motif chemokine receptor 2 (CCR2) overexpression enhanced the targeted migration and immunoregulatory potential of MSCs in response to C-C motif chemokine ligand 2 (CCL2) in vitro. Intravenously infusion of CCR2-engineered MSCs (MSCsCCR2) exhibited improved homing efficiencies to injured sites and lungs of diabetic mice. Accordingly, MSCCCR2 infusion inhibited monocyte infiltration, reshaped macrophage inflammatory properties, prompted the accumulation of regulatory T cells (Treg cells) in injured sites, and reshaped systemic immune responses via the lung and spleen in mouse diabetic wound models. In summary, CCR2-engineered MSCs restore immunological homeostasis to accelerate diabetic wound healing via their improved homing and immunoregulatory potentials in response to CCL2. Therefore, these findings provide a novel strategy to explore genetically engineered MSCs as tools to facilitate tissue repair in diabetic wounds.


Assuntos
Diabetes Mellitus Experimental , Células-Tronco Mesenquimais , Cicatrização , Animais , Diabetes Mellitus Experimental/terapia , Engenharia Genética , Homeostase , Humanos , Camundongos , Receptores CCR2
7.
Cell Mol Immunol ; 18(9): 2224-2235, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-32678310

RESUMO

Diabetes-associated periodontitis (DP) aggravates diabetic complications and increases mortality from diabetes. DP is caused by diabetes-enhanced host immune-inflammatory responses to bacterial insult. In this study, we found that persistently elevated CCL2 levels in combination with proinflammatory monocyte infiltration of periodontal tissues were closely related to DP. Moreover, inhibition of CCL2 by oral administration of bindarit reduced alveolar bone loss and increased periodontal epithelial thickness by suppressing periodontal inflammation. Furthermore, bindarit suppressed the infiltration of proinflammatory monocytes and altered the inflammatory properties of macrophages in the diabetic periodontium. This finding provides a basis for the development of an effective therapeutic approach for treating DP.


Assuntos
Quimiocina CCL2/antagonistas & inibidores , Diabetes Mellitus , Periodontite , Humanos , Indazóis/farmacologia , Macrófagos , Monócitos , Periodontite/complicações , Periodontite/tratamento farmacológico , Propionatos/farmacologia
8.
Bioact Mater ; 5(4): 1113-1126, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32743122

RESUMO

Periodontitis is caused by host immune-inflammatory response to bacterial insult. A high proportion of pro-inflammatory macrophages to anti-inflammatory macrophages leads to the pathogenesis of periodontitis. As stem cell-derived exosomes can modulate macrophage phenotype, dental pulp stem cell-derived exosomes (DPSC-Exo) can effectively treat periodontitis. In this study, we demonstrated that DPSC-Exo-incorporated chitosan hydrogel (DPSC-Exo/CS) can accelerate the healing of alveolar bone and the periodontal epithelium in mice with periodontitis. Gene Ontology (GO) term enrichment analysis showed that treatment with DPSC-Exo/CS ameliorated periodontal lesion by suppressing periodontal inflammation and modulating the immune response. Specifically, DPSC-Exo/CS facilitated macrophages to convert from a pro-inflammatory phenotype to an anti-inflammatory phenotype in the periodontium of mice with periodontitis, the mechanism of which could be associated with miR-1246 in DPSC-Exo. These results not only shed light on the therapeutic mechanism of DPSC-Exo/CS but also provide the basis for developing an effective therapeutic approach for periodontitis.

9.
J Periodontol ; 91(5): 705-714, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31471894

RESUMO

BACKGROUND: Hyperglycemic micro-environment induced by diabetes could regulate the response of periodontal tissues to pathogenic microorganisms in which disruption of autophagy lysosomal pathway (ALP) may participate. This study aimed to explore the mechanisms underlying how high glucose (HG) regulates ALP in gingival epithelial cells (GECs). METHODS: Human gingival tissues in healthy group (C), periodontitis group (P), diabetes group (DM), and diabetes + periodontitis group (DP) were collected and were applied to observe the fusion of autophagy and lysosome. Diabetic mouse model with periodontitis was established and RNA-seq was applied to investigate the expression of ALP-associated genes in gingival epithelium. To explore the key role of ATPase transmembrane v0 domain, subunit c (ATP6V0C) in the disruption of ALP by HG, human gingival epithelial cells (HGECs) were cultured in 5.5 mM/25 mM glucose medium for 48 hours and followed by Porphyromonas gingivalis stimulation for 0, 6, and 12 hours. HBLV-h-ATP6V0C was transfected in HGECs that were stimulated by 25 mM HG condition. RESULTS: Immunofluorescence double staining exhibited the disruption of ALP in human gingival epithelium in diabetes groups and HGECs under 25 mM glucose condition, accompanied with significantly downregulated lysosomal acidity. RNA-seq of mouse gingival epithelium screened out Atp6v0c. Compared with HGECs in normal culture medium, ATP6V0C expression and LC3-II/I expression ratio were significantly downregulated, with an upregulated expression of P62, IL-1ß in HGECs under HG condition. Over-expression of ATP6V0C rescued HG-induced disruption of ALP in HBLV-h-ATP6V0C transfected HGECs, with significantly upregulation of LC3-II/I and downregulation of P62, IL-1ß. CONCLUSION: ATP6V0C mediates HG-induced ALP disruption in HGECs, eventually exacerbates periodontal inflammation.


Assuntos
Autofagia , ATPases Vacuolares Próton-Translocadoras , Animais , Células Epiteliais , Gengiva , Glucose , Humanos , Lisossomos , Camundongos , Porphyromonas gingivalis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA