Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.050
Filtrer
1.
BMC Oral Health ; 24(1): 878, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095803

RÉSUMÉ

BACKGROUND: Effective treatments for the alveolar bone defect remain a major concern in dental therapy. The objectives of this study were to develop a fibrin and konjac glucomannan (KGM) composite hydrogel as scaffolds for the osteogenesis of nasal mucosa-derived ectodermal mesenchymal stem cells (EMSCs) for the regeneration of alveolar bone defect, and to investigate the osteogenesis-accelerating effects of black phosphorus nanoparticles (BPNs) embedded in the hydrogels. METHODS: Primary EMSCs were isolated from rat nasal mucosa and used for the alveolar bone recovery. Fibrin and KGM were prepared in different ratios for osteomimetic hydrogel scaffolds, and the optimal ratio was determined by mechanical properties and biocompatibility analysis. Then, the optimal hydrogels were integrated with BPNs to obtain BPNs/fibrin-KGM hydrogels, and the effects on osteogenic EMSCs in vitro were evaluated. To explore the osteogenesis-enhancing effects of hydrogels in vivo, the BPNs/fibrin-KGM scaffolds combined with EMSCs were implanted to a rat model of alveolar bone defect. Micro-computed tomography (CT), histological examination, real-time quantitative polymerase chain reaction (RT-qPCR) and western blot were conducted to evaluate the bone morphology and expression of osteogenesis-related genes of the bone regeneration. RESULTS: The addition of KGM improved the mechanical properties and biodegradation characteristics of the fibrin hydrogels. In vitro, the BPNs-containing compound hydrogel was proved to be biocompatible and capable of enhancing the osteogenesis of EMSCs by upregulating the mineralization and the activity of alkaline phosphatase. In vivo, the micro-CT analysis and histological evaluation demonstrated that rats implanted EMSCs-BPNs/fibrin-KGM hydrogels exhibited the best bone reconstruction. And compared to the model group, the expression of osteogenesis genes including osteopontin (Opn, p < 0.0001), osteocalcin (Ocn, p < 0.0001), type collagen (Col , p < 0.0001), bone morphogenetic protein-2 (Bmp2, p < 0.0001), Smad1 (p = 0.0006), and runt-related transcription factor 2 (Runx2, p < 0.0001) were all significantly upregulated. CONCLUSIONS: EMSCs/BPNs-containing fibrin-KGM hydrogels accelerated the recovery of the alveolar bone defect in rats by effectively up-regulating the expression of osteogenesis-related genes, promoting the formation and mineralisation of bone matrix.


Sujet(s)
Régénération osseuse , Fibrine , Hydrogels , Mannanes , Cellules souches mésenchymateuses , Ostéogenèse , Phosphore , Rat Sprague-Dawley , Structures d'échafaudage tissulaires , Animaux , Régénération osseuse/effets des médicaments et des substances chimiques , Rats , Mannanes/pharmacologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Ostéogenèse/effets des médicaments et des substances chimiques , Microtomographie aux rayons X , Nanoparticules , Muqueuse nasale , Processus alvéolaire , Mâle , Protéine morphogénétique osseuse de type 2 , Sous-unité alpha 1 du facteur CBF , Ostéocalcine
2.
Physiol Res ; 73(3): 415-426, 2024 07 17.
Article de Anglais | MEDLINE | ID: mdl-39027958

RÉSUMÉ

Recent research has identified that miR-539-3p impedes chondrogenic differentiation, yet its specific role and underlying mechanisms in childhood-onset osteoarthritis (OA) remain unclear. This study found that miR-539-3p levels were considerably lower in cartilage samples derived from childhood-onset OA patients compared to the control group. Enhancing miR-539-3p expression or suppressing RUNX2 expression notably reduced apoptosis, inflammation, and extracellular matrix (ECM) degradation in OA chondrocytes. In contrast, reducing miR-539-3p or increasing RUNX2 had the opposite effects. RUNX2 was confirmed as a direct target of miR-539-3p. Further experiments demonstrated that miR-539-3p targeting RUNX2 effectively lessened apoptosis, inflammation, and ECM degradation in OA chondrocytes, accompanied by changes in key molecular markers like reduced caspase-3 and matrix etallopeptidase 13 (MMP-13) levels, and increased B-cell lymphoma 2 (Bcl-2) and collagen type X alpha 1 chain (COL2A1). This study underscores the pivotal role of miR-539-3p in alleviating inflammation and ECM degradation in childhood-onset OA through targeting RUNX2, offering new insights for potential therapeutic strategies against this disease.


Sujet(s)
Apoptose , Chondrocytes , Sous-unité alpha 1 du facteur CBF , Matrice extracellulaire , microARN , Arthrose , Humains , microARN/métabolisme , microARN/génétique , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/anatomopathologie , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/génétique , Enfant , Mâle , Femelle , Cellules cultivées , Adolescent
3.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(3): 304-312, 2024 Jun 01.
Article de Anglais, Chinois | MEDLINE | ID: mdl-39049649

RÉSUMÉ

OBJECTIVES: This study aims to investigate the influence of glucose regulated protein (GRP) 78 on osteoblast differentiation in periodontal ligament fibroblasts (PDLFs) under cyclic mechanical stretch and determine the underlying mechanism. METHODS: FlexCell 5000 cell mechanical device was applied to simulate the stress environment of orthodontic teeth. GRP78High and GRP78Low subpopulation were obtained by flow sorting. Gene transfection was performed to knockdown GRP78 and c-Src expression and overexpress c-Src. Western blot analysis was used to detect the protein expression of Runt-related gene 2 (RUNX2), Osterix, osteocalcin (OCN), and osteopontin (OPN). Immunoprecipitation assay was used to determine the interaction of GRP78 with c-Src. The formation of cellular mineralized nodules was determined by alizarin red staining. RESULTS: GRP78 was heterogeneously expressed in PDLFs, and GRP78High and GRP78Low subpopulations were obtained by flow sorting. The osteogenic differentiation ability and phosphorylation level of c-Src kinase in the GRP78High subpopulation were significantly increased compared with those in GRP78Low subpopulation after cyclic mechanical stretch (P<0.05). GRP78 interacted with c-Src in PDLFs. The overexpression c-Src group showed significantly increased osteogenic differentiation ability than the vector group (P<0.05), and the sic-Src group showed significantly decreased osteogenic differentiation ability (P<0.05) after cyclic mechanical stretch. CONCLUSIONS: GRP78 upregulates c-Src expression by interacting with c-Src kinase and promotes osteogenic differentiation under cyclic mechanical stretch in PDLFs.


Sujet(s)
Différenciation cellulaire , Chaperonne BiP du réticulum endoplasmique , Fibroblastes , Protéines du choc thermique , Ostéoblastes , Ostéogenèse , Desmodonte , Transduction du signal , Contrainte mécanique , Desmodonte/métabolisme , Desmodonte/cytologie , Chaperonne BiP du réticulum endoplasmique/métabolisme , Ostéoblastes/métabolisme , Fibroblastes/métabolisme , Humains , Protéines du choc thermique/métabolisme , Ostéopontine/métabolisme , Ostéocalcine/métabolisme , src-Family kinases/métabolisme , Phosphorylation , Sous-unité alpha 1 du facteur CBF/métabolisme , CSK tyrosine-protein kinase/métabolisme
4.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(7): 1227-1235, 2024 Jul 20.
Article de Chinois | MEDLINE | ID: mdl-39051068

RÉSUMÉ

OBJECTIVE: To investigate the role of high-mobility group AT-hook 2 (HMGA2) in osteogenic differentiation of adipose-derived mesenchymal stem cells (ADSCs) and the effect of Hmga2 knockdown for promoting bone defect repair. METHODS: Bioinformatics studies using the GEO database and Rstudio software identified HMGA2 as a key factor in adipogenic-osteogenic differentiation balance of ADSCs. The protein-protein interaction network of HMGA2 in osteogenic differentiation was mapped using String and visualized with Cytoscape to predict the downstream targets of HMGA2. Primary mouse ADSCs (mADSCs) were transfected with Hmga2 siRNA, and the changes in osteogenic differentiation of the cells were evaluated using alkaline phosphatase staining and Alizarin red S staining. The expressions of osteogenic markers Runt-related transcription factor 2 (RUNX2), osteopontin (OPN), and osteocalcein (OCN) in the transfected cells were detected using RT-qPCR and Western blotting. In a mouse model of critical-sized calvarial defects, mADSCs with Hmga2-knockdown were transplanted into the defect, and bone repair was evaluated 6 weeks later using micro-CT scanning and histological staining. RESULTS: GEO database analysis showed that HMGA2 expression was upregulated during adipogenic differentiation of ADSCs. Protein-protein interaction network analysis suggested that the potential HMGA2 targets in osteogenic differentiation of ADSCs included SMAD7, CDH1, CDH2, SNAI1, SMAD9, IGF2BP3, and ALDH1A1. In mADSCs, Hmga2 knockdown significantly upregulated the expressions of RUNX2, OPN, and OCN and increased cellular alkaline phosphatase activity and calcium deposition. In a critical-sized calvarial defect model, transplantation of mADSCs with Hmga2 knockdown significantly promoted new bone formation. CONCLUSION: HMGA2 is a crucial regulator of osteogenic differentiation in ADSCs, and Hmga2 knockdown significantly promotes osteogenic differentiation of ADSCs and accelerates ADSCs-mediated bone defect repair in mice.


Sujet(s)
Différenciation cellulaire , Protéine HMGA2 , Cellules souches mésenchymateuses , Ostéogenèse , Animaux , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Protéine HMGA2/génétique , Protéine HMGA2/métabolisme , Souris , Tissu adipeux/cytologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Petit ARN interférent/génétique , Techniques de knock-down de gènes , Adipogenèse/génétique
5.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000154

RÉSUMÉ

Putatively, tooth agenesis was attributed to the initiation failure of tooth germs, though little is known about the histological and molecular alterations. To address if constitutively active FGF signaling is associated with tooth agenesis, we activated Fgf8 in dental mesenchyme with Osr-cre knock-in allele in mice (Osr2-creKI; Rosa26R-Fgf8) and found incisor agenesis and molar microdontia. The cell survival assay showed tremendous apoptosis in both the Osr2-creKI; Rosa26R-Fgf8 incisor epithelium and mesenchyme, which initiated incisor regression from cap stage. In situ hybridization displayed vanished Shh transcription, and immunostaining exhibited reduced Runx2 expression and enlarged mesenchymal Lef1 domain in Osr2-creKI; Rosa26R-Fgf8 incisors, both of which were suggested to enhance apoptosis. In contrast, Osr2-creKI; Rosa26R-Fgf8 molar germs displayed mildly suppressed Shh transcription, and the increased expression of Ectodin, Runx2 and Lef1. Although mildly smaller than WT controls prenatally, the Osr2-creKI; Rosa26R-Fgf8 molar germs produced a miniature tooth with impaired mineralization after a 6-week sub-renal culture. Intriguingly, the implanted Osr2-creKI; Rosa26R-Fgf8 molar germs exhibited delayed odontoblast differentiation and accelerated ameloblast maturation. Collectively, the ectopically activated Fgf8 in dental mesenchyme caused incisor agenesis by triggering incisor regression and postnatal molar microdontia. Our findings reported tooth agenesis resulting from the regression from the early bell stage and implicated a correlation between tooth agenesis and microdontia.


Sujet(s)
Facteur de croissance fibroblastique de type 8 , Incisive , Mésoderme , Molaire , Animaux , Facteur de croissance fibroblastique de type 8/génétique , Facteur de croissance fibroblastique de type 8/métabolisme , Souris , Incisive/malformations , Incisive/métabolisme , Mésoderme/métabolisme , Mésoderme/anatomopathologie , Molaire/malformations , Molaire/métabolisme , Anodontie/génétique , Anodontie/métabolisme , Anodontie/anatomopathologie , Apoptose , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Facteur de transcription LEF-1/métabolisme , Facteur de transcription LEF-1/génétique , Sous-unité alpha 1 du facteur CBF/génétique , Sous-unité alpha 1 du facteur CBF/métabolisme , Transduction du signal , Régulation de l'expression des gènes au cours du développement , Odontogenèse/génétique , Souris transgéniques
6.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-39000419

RÉSUMÉ

The search for the molecular markers of osteoporosis (OP), based on the analysis of differential deoxyribonucleic acid (DNA) methylation in bone cells and peripheral blood cells, is promising for developments in the field of the early diagnosis and targeted therapy of the disease. The Runt-related transcription factor 2 (RUNX2) gene is one of the key genes of bone metabolism, which is of interest in the search for epigenetic signatures and aberrations associated with the risk of developing OP. Based on pyrosequencing, the analysis of the RUNX2 methylation profile from a pool of peripheral blood cells in men and women over 50 years of age of Russian ethnicity from the Volga-Ural region of Russia was carried out. The level of DNA methylation in three CpG sites of the RUNX2 gene was assessed and statistically significant hypomethylation was revealed in all three studied CpG sites in men (U = 746.5, p = 0.004; U = 784, p = 0.01; U = 788.5, p = 0.01, respectively) and in one CpG site in women (U = 537, p = 0.03) with primary OP compared with control. In the general sample, associations were preserved for the first CpG site (U = 2561, p = 0.0001766). The results were obtained for the first time and indicate the existence of potentially new epigenetic signatures of RUNX2 in individuals with OP.


Sujet(s)
Marqueurs biologiques , Sous-unité alpha 1 du facteur CBF , Ilots CpG , Méthylation de l'ADN , Ostéoporose , Humains , Sous-unité alpha 1 du facteur CBF/génétique , Mâle , Femelle , Ostéoporose/génétique , Adulte d'âge moyen , Ilots CpG/génétique , Sujet âgé , Épigenèse génétique
7.
Shanghai Kou Qiang Yi Xue ; 33(2): 130-134, 2024 Apr.
Article de Chinois | MEDLINE | ID: mdl-39005087

RÉSUMÉ

PURPOSE: To investigate the therapeutic effect of atorvastatin on alveolar bone defect model in rats, and to observe the effect of atorvastatin on Wnt/ß-catenin. METHODS: Thirty rats were randomly divided into normal group (group N), model group (group M) and atorvastatin administration group (group ATV). Except group N, bone defects were made in other rats' alveolar bone to construct alveolar bone defect model. After successful modeling, 20 mg/kg atorvastatin suspension was administered by gavage in group ATV, and the same amount of sodium carboxymethyl cellulose solution was administered by gavage in group N and group M for twenty-one days. After the last administration, tail vein blood was collected to detect the concentrations of serum osteoprotegerin (OPG), alkaline phosphatase (ALP) and osteocalcin (BPG). H-E staining was used to observe the pathological changes of maxillary defect area, and lane Sandhu score was performed. Tartrate resistant acid phosphatase(TRAP) staining was used to detect the number of osteoclasts in the defect area. Real time fluorescence quantitative PCR(RT-qPCR) and Western blot(WB) were used to detect Wnt, ß-catenin and Runx2 mRNA protein expression. Statistical analysis was performed with SPSS 23.0 software package. RESULTS: Compared with group N, the concentrations of OPG, ALP, BGP and Lane Sandhu score in group M decreased, and the number of osteoclasts increased. Compared with group M, the concentrations of OPG, ALP and BGP and lane Sandhu score in group ATV increased, and the number of osteoclasts decreased. After H-E staining, the amount of bone formation in maxillary defect area in group N was more,there was fewer bone tissues in the defect area in group M, the amount of bone tissues in the defect area increased in group ATV. Compared with group N, Wnt, ß-catenin and Runx2 mRNA protein decreased. Compared with group M, Wnt, ß-catenin and Runx2 mRNA protein expression increased. CONCLUSIONS: Atorvastatin can promote the healing of alveolar bone defect and accelerate bone reconstruction in rat models. This effect may be related to the activation of Wnt/ß-catenin signaling pathway.


Sujet(s)
Phosphatase alcaline , Atorvastatine , Ostéocalcine , Ostéoprotégérine , Voie de signalisation Wnt , bêta-Caténine , Animaux , Atorvastatine/pharmacologie , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Rats , Ostéoprotégérine/métabolisme , Ostéoprotégérine/génétique , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Ostéocalcine/métabolisme , Ostéocalcine/génétique , Ostéocalcine/sang , Phosphatase alcaline/métabolisme , Phosphatase alcaline/sang , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Processus alvéolaire/effets des médicaments et des substances chimiques , Processus alvéolaire/métabolisme
8.
Shanghai Kou Qiang Yi Xue ; 33(2): 135-140, 2024 Apr.
Article de Chinois | MEDLINE | ID: mdl-39005088

RÉSUMÉ

PURPOSE: To investigate the effect of TNF-α on osteogenic differentiation of stem cells from human exfoliated deciduous teeth (SHED), and to analyze the changes of ERK1/2-Runx2 signaling pathway in the regulation process. METHODS: SHED cells were isolated and cultured from normal deciduous permanent teeth of healthy children aged 6-8 years old, and the third passage of SHED cells were taken and divided into control group (osteogenic inducer culture), observation group (osteogenic inducer and TNF-α co-culture) and agonist group (osteogenic inducer, TNF-α and ERK pathway agonist co-culture). The osteogenic differentiation was determined by alizarin red staining. The protein expression levels of Osterix, OPN, ERK1/2, pERK1/2 and Runx2 in SHED cells were determined by Western blot. The expressions of Osterix, OPN, ERK1/2, pERK1/2 and Runx2 mRNA were detected by qRT-PCR. Statistical analysis was performed with SPSS 26.0 software package. RESULTS: Comparison of osteogenic differentiation ability of the three groups of cells showed that red-brown mineralized nodules were observed in the three groups of cells. Compared among the three groups, the control group had the most mineralized nodules, followed by the activation group, and the observation group had the least mineralized nodules. Compared with the control group, the expression levels of Osterix and OPN protein and mRNA in the observation group and the agonist group were significantly decreased, while the expression levels of Osterix and OPN protein and mRNA in the agonist group were significantly higher than those in the observation group. There was no significant difference in the expression levels of ERK1/2 protein and mRNA among the three groups, while the expression levels of pERK1/2 and Runx2 protein and mRNA in the observation group and the agonist group were significantly higher than those in the control group, and the expression levels of pERK1/2 and Runx2 protein and mRNA in the agonist group were significantly higher than those in the observation group. CONCLUSIONS: TNF-α can inhibit osteogenic differentiation of SHED cells, which may be related to the inhibition of ERK1/2-Runx2 signaling pathway.


Sujet(s)
Différenciation cellulaire , Sous-unité alpha 1 du facteur CBF , Système de signalisation des MAP kinases , Ostéogenèse , Dent de lait , Facteur de nécrose tumorale alpha , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Ostéogenèse/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Enfant , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Dent de lait/cytologie , Dent de lait/métabolisme , Facteur de transcription Sp7/métabolisme , Facteur de transcription Sp7/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Cellules souches/métabolisme , Cellules souches/cytologie , Cellules cultivées
9.
Clin Oral Investig ; 28(8): 426, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38992200

RÉSUMÉ

OBJECTIVES: To assess the short-term efficacy of multiple sessions of antimicrobial photodynamic therapy (aPDT), light-emitting-diode (LED) photobiomodulation, and topical ozone therapy applications following surgical regenerative treatments on clinical parameters, patient-centered outcomes, and mRNA expression levels of VEGF, IL-6, RunX2, Nell-1, and osterix in gingival crevicular fluid samples in patients with stage III/IV, grade C periodontitis. MATERIALS AND METHODS: Forty-eight systemically healthy patients were assigned into four groups to receive adjunctive modalities with regenerative periodontal surgical treatment. A 970 ± 15 nm diode laser plus indocyanine-green for aPDT group, a 626 nm LED for photobiomodulation group, and topical gaseous ozone were applied at 0, 1, 3, and 7 postoperative days and compared to control group. The clinical periodontal parameters, early wound healing index (EHI), and postoperative patients' morbidity were evaluated. The mRNA levels of biomarkers were assessed by real-time polymerase chain reaction. RESULTS: No significant difference in the clinical parameters except gingival recession (GR) was identified among the groups. For group-by-time interactions, plaque index (PI) and probing pocket depths (PD) showed significant differences (p = 0.034; p = 0.022). In sites with initial PD > 7 mm, significant differences were observed between control and photobiomodulation groups in PD (p = 0.011), between control and aPDT, and control and photobiomodulation groups in CAL at 6-month follow-up (p = 0.007; p = 0.022). The relative osterix mRNA levels showed a statistically significant difference among the treatment groups (p = 0.014). CONCLUSIONS: The additional applications of aPDT and LED after regenerative treatment of stage III/IV grade C periodontitis exhibited a more pronounced beneficial effect on clinical outcomes in deep periodontal pockets.


Sujet(s)
Lasers à semiconducteur , Photothérapie de faible intensité , Ozone , Photothérapie dynamique , Humains , Photothérapie dynamique/méthodes , Mâle , Femelle , Ozone/usage thérapeutique , Adulte , Photothérapie de faible intensité/méthodes , Lasers à semiconducteur/usage thérapeutique , Résultat thérapeutique , Adulte d'âge moyen , Parodontite/thérapie , Vert indocyanine/usage thérapeutique , Association thérapeutique , Réaction de polymérisation en chaine en temps réel , Exsudat gingival , Marqueurs biologiques , Photosensibilisants/usage thérapeutique , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Indice parodontal , Interleukine-6 , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Sous-unité alpha 1 du facteur CBF , Facteur de transcription Sp7
10.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 663-671, 2024 Jul 09.
Article de Chinois | MEDLINE | ID: mdl-38949134

RÉSUMÉ

Objective: To investigate the characteristics of extracellular matrix vesicle mimetics prepared by mechanical extrusion and their effects on the cell viability and osteogenic differentiation potential of human periodontal ligament stem cells (PDLSC). Methods: PDLSC derived extracellular matrix vesicles were prepared by collagenase digestion, while the cell derived vesicle mimetics were simulated by mechanical extrusion. The obtained extracellular matrix vesicles and parental cell derived vesicle mimetics were divided into 4 groups: matrix vesicles derived from PDLSC cultured in basic medium for 7 days (PDLSC matrix vesicles, MVs), vesicle mimetics derived from PDLSC cultured in basic medium for 7 days (PDLSC vesicle mimetics, CVMs), matrix vesicles derived from PDLSC cultured in osteogenic inducing medium for 7 days (osteogenic-induced PDLSC matrix vesicles, O-MVs) and vesicle mimetics derived from PDLSC cultured in osteogenic inducing medium for 7 days (osteogenic-induced PDLSC vesicle mimetics, O-CVMs). Vesicles morphologies and sizes were observed by transmission electron microscopy and nanoparticle tracking analysis. Vesicles uptake was detected by immunofluorescence. With PDLSC as the control group, the effects of vesicles on the viability of PDLSC were detected by cell activity assay (cell counting kit-8), and the effects of vesicles on the osteogenic differentiation potential of PDLSC were detected by alizarin red staining and Western blotting. Results: Vesicles in MVs, O-MVs, CVMs and O-CVMs were all observed with a round structure (size 50-250 nm), and could be taken up by PDLSC without affecting the cell viability. Under osteogenic inducing conditions, PDLSC incubated with O-MVs or O-CVMs could produce more mineralized nodules than those in the control group (PDLSC). MVs, O-MVs, CVMs and O-CVMs could promote the expression of osteogenic-related proteins in PDLSC. PDLSC in group O-CVMs showed significant higher expressions of osteogenic-related proteins, including alkaline phosphatase (ALP) (1.571±0.348), osteopontin (OPN) (1.827±0.627) and osteocalcin (OCN) (1.798±0.537) compared to MVs (ALP: 1.156±0.170, OPN: 1.260±0.293, OCN: 1.286±0.302) (P<0.05). Compared to CMVs-incubated PDLSC, O-CVMs-incubated PDLSC expressed more Runt-related transcription factor 2 (1.632±0.455 vs 1.176±0.128) and OPN (1.827±0.627 vs 1.428±0.427) (P<0.05). Moreover, there was no significant difference in the expression levels of osteoblast-related proteins in PDLSC cultured with MVs, O-MVs and CVMs (P>0.05). Conclusions: The vesicle mimetics prepared by mechanical extrusion method are similar in shape and size to the extracellular matrix vesicles. MVs, O-MVs, CVMs and O-CVMs do not affect the cell viability of PDLSC, and can promote the osteogenic differentiation potential of PDLSC to a certain extent.


Sujet(s)
Différenciation cellulaire , Matrice extracellulaire , Vésicules extracellulaires , Ostéogenèse , Humains , Matrice extracellulaire/métabolisme , Vésicules extracellulaires/métabolisme , Cellules souches/cytologie , Phosphatase alcaline/métabolisme , Desmodonte/cytologie , Desmodonte/métabolisme , Ostéocalcine/métabolisme , Ostéopontine/métabolisme , Sous-unité alpha 1 du facteur CBF/métabolisme
11.
Lasers Med Sci ; 39(1): 174, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38969931

RÉSUMÉ

PURPOSE: Laser irradiation activates a range of cellular processes in the periodontal components and promotes tissue repair. However, its effect on osteogenic differentiation of human cementoblast lineage cells remains unclear. This study aimed to examine the effects of high-frequency semiconductor laser irradiation on the osteogenic differentiation of human cementoblast lineage (HCEM) cells. METHODS: HCEM cells were cultured to reach 80% confluence and irradiated with a gallium-aluminum-arsenide (Ga-Al-As) semiconductor laser with a pulse width of 200 ns and wavelength of 910 at a dose of 0-2.0 J/cm2. The outcomes were assessed by analyzing the mRNA levels of alkaline phosphatase (ALP), runt-related transcription factor 2 (RUNX2), and type I collagen (COLL1) using real-time polymerase chain reaction (PCR) analysis 24 h after laser irradiation. Cell mineralization was evaluated using ALP activity, calcium deposition, and Alizarin Red staining. RESULTS: The laser-irradiated HCEM cells showed significantly enhanced gene expression levels of ALP, RUNX2, and COLL1 as well as ALP activity and calcium concentration in the culture medium compared with the non-irradiated cells. In addition, enhanced calcification deposits were confirmed in the laser-irradiated group compared with the non-irradiated group at 21 and 28 days after the induction of osteogenic differentiation. CONCLUSION: High-frequency semiconductor laser irradiation enhances the osteogenic differentiation potential of cultured HCEM cells, underscoring its potential utility for periodontal tissue regeneration.


Sujet(s)
Différenciation cellulaire , Cément dentaire , Lasers à semiconducteur , Ostéogenèse , Humains , Lasers à semiconducteur/usage thérapeutique , Différenciation cellulaire/effets des radiations , Ostéogenèse/effets des radiations , Cément dentaire/effets des radiations , Cément dentaire/cytologie , Phosphatase alcaline/métabolisme , Cellules cultivées , Photothérapie de faible intensité/méthodes , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Collagène de type I/génétique , Collagène de type I/métabolisme
12.
Cell Signal ; 121: 111294, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38996954

RÉSUMÉ

BACKGROUND: Osteoporosis (OP) is a prevalent disease associated with age, and one of the primary pathologies is the defect of osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). This study aimed to elucidate whether Nuclear Receptor Binding SET Domain Protein 2 (NSD2) transcriptionally regulates osteogenic differentiation of BMSCs in osteoporosis. METHODS: Identification of human BMSCs (hBMSCs) in vitro was measured by flow cytometry. Osteogenesis of hBMSCs in vitro was measured by Alizarin Red and Alkaline Phosphatase staining. The protein levels of H3K36me1/2/3, NSD2, and Hoxa2 were measured by western blotting. The mRNA levels of NSD2, Runx2, and BSP were measured by qPCR. The role of NSD2 in the osteogenic differentiation of BMSCs was further identified by silencing NSD2 via shRNA or overexpression of NSD2 via lentivirus transfection. The interactions of NSD2, H3K36me2 and Hoxa2 were identified via chromatin immunoprecipitation (ChIP). Luciferase reporting analysis was employed to confirm that NSD2 regulated the transcriptional activity of Hoxa2. Ovariectomized (OVX) was performed on mice to construct osteoporosis (OP) model. Subsequently, the bone mass was assessed by micro computed tomography (micro-CT) scan. RESULTS: During the osteogenesis of OP-derived hBMSCs, the levels of NSD2 and H3K36me2 significantly increased in 14 days of osteogenic induction. Inhibition of NSD2 via shRNA increased the RUNX2 and BSP expression of hBMSCs, while overexpression of NSD2 decreased RUNX2 and BSP expression of hBMSCs. ChIP analysis indicated NSD2-mediated H3K36me2 reduced the osteogenic differentiation of hBMSCs by regulating the osteogenic inhibitor Hoxa2. Accordingly, inhibition of NSD2 in vivo via tail vein injection of LV-shNSD2 lentivirus greatly alleviated OVX-induced osteoporosis in mice. CONCLUSION: We demonstrated that NSD2 inhibited the osteogenic differentiation in hBMSCs by transcriptionally downregulating Hoxa2 via H3K36me2 dimethylation. Inhibition of NSD2 effectively attenuated bone loss in murine osteoporosis and NSD2 is a promising target for clinical treatment of osteoporosis.


Sujet(s)
Différenciation cellulaire , Histone-lysine N-methyltransferase , Protéines à homéodomaine , Cellules souches mésenchymateuses , Ostéogenèse , Ostéoporose , Cellules souches mésenchymateuses/métabolisme , Ostéoporose/métabolisme , Ostéoporose/anatomopathologie , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Humains , Animaux , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Souris , Femelle , Histone/métabolisme , Protéines de répression/métabolisme , Sous-unité alpha 1 du facteur CBF/métabolisme , Souris de lignée C57BL , Cellules cultivées
13.
Int J Mol Sci ; 25(14)2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39062933

RÉSUMÉ

This study investigates the impact of acetylsalicylic acid (ASA), also known as aspirin, on adipose tissue-derived stem cells (ASCs), aiming to elucidate its dose-dependent effects on morphology, viability, proliferation, and osteogenic differentiation. Isolated and characterized human ASCs were exposed to 0 µM, 100 µM, 200 µM, 400 µM, 800 µM, 1000 µM, 10,000 µM, and 16,000 µM of ASA in vitro. Cell morphology, viability, and proliferation were evaluated with fluorescent live/dead staining, alamarBlue viability reagent, and CyQUANT® cell proliferation assay, respectively. Osteogenic differentiation under stimulation with 400 µM or 1000 µM of ASA was assessed with alizarin red staining and qPCR of selected osteogenic differentiation markers (RUNX2, SPP1, ALPL, BGLAP) over a 3- and 21-day-period. ASA doses ≤ 1000 µM showed no significant impact on cell viability and proliferation. Live/dead staining revealed a visible reduction in viable cell confluency for ASA concentrations ≥ 1000 µM. Doses of 10,000 µM and 16,000 µM of ASA exhibited a strong cytotoxic and anti-proliferative effect in ASCs. Alizarin red staining revealed enhanced calcium accretion under the influence of ASA, which was macro- and microscopically visible and significant for 1000 µM of ASA (p = 0.0092) in quantification if compared to osteogenic differentiation without ASA addition over a 21-day-period. This enhancement correlated with a more pronounced upregulation of osteogenic markers under ASA exposure (ns). Our results indicate a stimulatory effect of 1000 µM of ASA on the osteogenic differentiation of ASCs. Further research is needed to elucidate the precise molecular mechanisms underlying this effect; however, this discovery suggests promising opportunities for enhancing bone tissue engineering with ASCs as cell source.


Sujet(s)
Tissu adipeux , Acide acétylsalicylique , Différenciation cellulaire , Prolifération cellulaire , Survie cellulaire , Ostéogenèse , Cellules souches , Humains , Acide acétylsalicylique/pharmacologie , Ostéogenèse/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Tissu adipeux/cytologie , Tissu adipeux/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules souches/effets des médicaments et des substances chimiques , Cellules souches/cytologie , Cellules souches/métabolisme , Cellules cultivées , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Ostéopontine/métabolisme , Ostéopontine/génétique , Ostéocalcine/métabolisme , Ostéocalcine/génétique , Phosphatase alcaline/métabolisme , Femelle , Adulte
14.
J Contemp Dent Pract ; 25(4): 313-319, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38956844

RÉSUMÉ

AIMS: This study aims to assess the synergistic effect of utilizing a bioceramic sealer, NeoPutty, with photobiomodulation (PBM) on dental pulp stem cells (DPSCs) for odontogenesis. MATERIALS AND METHODS: Dental pulp stem cells were collected from 10 premolars extracted from healthy individuals. Dental pulp stem cells were characterized using an inverted-phase microscope to detect cell shape and flow cytometry to detect stem cell-specific surface antigens. Three experimental groups were examined: the NP group, the PBM group, and the combined NP and PBM group. A 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) experiment was conducted to assess the viability of DPSCs. The odontogenic differentiation potential was analyzed using Alizarin red staining, RT-qPCR analysis of odontogenic genes DMP-1, DSPP, and alkaline phosphatase (ALP), and western blot analysis for detecting BMP-2 and RUNX-2 protein expression. An analysis of variance (ANOVA) followed by a post hoc t-test was employed to examine and compare the mean values of the results. RESULTS: The study showed a notable rise in cell viability when NP and PBM were used together. Odontogenic gene expression and the protein expression of BMP-2 and RUNX-2 were notably increased in the combined group. The combined effect of NeoPutty and PBM was significant in enhancing the odontogenic differentiation capability of DPSCs. CONCLUSION: The synergistic effect of NeoPutty and PBM produced the most positive effect on the cytocompatibility and odontogenic differentiation potential of DPSCs. CLINICAL SIGNIFICANCE: Creating innovative regenerative treatments to efficiently and durably repair injured dental tissues. How to cite this article: Alshawkani HA, Mansy M, Al Ankily M, et al. Regenerative Potential of Dental Pulp Stem Cells in Response to a Bioceramic Dental Sealer and Photobiomodulation: An In Vitro Study. J Contemp Dent Pract 2024;25(4):313-319.


Sujet(s)
Protéine morphogénétique osseuse de type 2 , Différenciation cellulaire , Pulpe dentaire , Photothérapie de faible intensité , Odontogenèse , Cellules souches , Pulpe dentaire/cytologie , Humains , Cellules souches/effets des médicaments et des substances chimiques , Photothérapie de faible intensité/méthodes , Différenciation cellulaire/effets des médicaments et des substances chimiques , Odontogenèse/effets des médicaments et des substances chimiques , Produits d'obturation des canaux radiculaires/pharmacologie , Phosphatase alcaline/métabolisme , Techniques in vitro , Survie cellulaire/effets des médicaments et des substances chimiques , Régénération/effets des médicaments et des substances chimiques , Céramiques , Protéines de la matrice extracellulaire , Cellules cultivées , Sous-unité alpha 1 du facteur CBF , Sialoglycoprotéines , Phosphoprotéines
15.
Sci Rep ; 14(1): 16323, 2024 07 15.
Article de Anglais | MEDLINE | ID: mdl-39009669

RÉSUMÉ

Vascular calcification, which is a major complication of diabetes mellitus, is an independent risk factor for cardiovascular disease. Osteogenic differentiation of vascular smooth muscle cells (VSMCs) is one of the key mechanisms underlying vascular calcification. Emerging evidence suggests that macrophage-derived extracellular vesicles (EVs) may be involved in calcification within atherosclerotic plaques in patients with diabetes mellitus. However, the role of macrophage-derived EVs in the progression of vascular calcification is largely unknown. In this study, we investigated whether macrophage-derived EVs contribute to the osteogenic differentiation of VSMCs under high glucose conditions. We isolated EVs that were secreted by murine peritoneal macrophages under normal glucose (EVs-NG) or high glucose (EVs-HG) conditions. miRNA array analysis in EVs from murine macrophages showed that miR-17-5p was significantly increased in EVs-HG compared with EVs-NG. Prediction analysis with miRbase identified transforming growth factor ß receptor type II (TGF-ß RII) as a potential target of miR-17-5p. EVs-HG as well as miR-17-5p overexpression with lipid nanoparticles inhibited the gene expression of Runx2, and TGF-ß RII. Furthermore, we demonstrated that VSMCs transfected with miR-17-5p mimic inhibited calcium deposition. Our findings reveal a novel role of macrophage-derived EVs in the negative regulation of osteogenic differentiation in VSMCs under high glucose conditions.


Sujet(s)
Différenciation cellulaire , Vésicules extracellulaires , Glucose , microARN , Muscles lisses vasculaires , Myocytes du muscle lisse , Ostéogenèse , Transduction du signal , Facteur de croissance transformant bêta , microARN/génétique , microARN/métabolisme , Animaux , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/cytologie , Glucose/pharmacologie , Glucose/métabolisme , Ostéogenèse/effets des médicaments et des substances chimiques , Ostéogenèse/génétique , Facteur de croissance transformant bêta/métabolisme , Souris , Myocytes du muscle lisse/métabolisme , Vésicules extracellulaires/métabolisme , Calcification vasculaire/métabolisme , Calcification vasculaire/génétique , Calcification vasculaire/anatomopathologie , Récepteur de type II du facteur de croissance transformant bêta/métabolisme , Récepteur de type II du facteur de croissance transformant bêta/génétique , Mâle , Souris de lignée C57BL , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique
16.
Indian J Pharmacol ; 56(3): 198-205, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-39078184

RÉSUMÉ

OBJECTIVES: Single-nucleotide polymorphism (SNP) codes for multiple amino acids, impacting protein functions and disease prognosis. Runt-related transcription factor-2 (RUNX2), a transcription factor linked to osteoblast differentiation, regulates cell proliferation in endothelium and osteoblastic cells. Understanding Runx2's role in nonosseous tissues is rapidly advancing. This study aims to identify harmful SNPs of the RUNX2 gene that may alter disease susceptibility using computational techniques. METHODS: The study uses various in silico methods to identify nonsynonymous SNPs (nsSNPs) of the RUNX2 gene, which could potentially alter protein structure and functions, with further analyses by I-Mutant, ConSurf, Netsurf 3.0, GeneMANIA, and Have (y)Our Protein Explained. RESULTS: Six missense nsSNPs were identified as potentially harmful, disease-causing, and damaging. Four were found to be unstable, while five were conserved. All six nsSNPs had a coiled secondary structure. Five nsSNPs were found to be destabilized. CONCLUSION: The RUNX2 gene's deleterious missense nsSNPs were identified by this study, and they may be exploited in future experimental studies. These high-risk nsSNPs might be considered target molecules in therapeutic and diagnostic therapies in teeth and bone development.


Sujet(s)
Simulation numérique , Sous-unité alpha 1 du facteur CBF , Mutation faux-sens , Polymorphisme de nucléotide simple , Sous-unité alpha 1 du facteur CBF/génétique , Humains , Biologie informatique , Prédisposition génétique à une maladie
17.
Int Immunopharmacol ; 138: 112573, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38971108

RÉSUMÉ

BACKGROUND: Tianhe Zhuifeng Gao (TZG) is an authorized Chinese patent drug with satisfying clinical efficacy, especially for RA patients with cold-dampness syndrome. However, its underlying pharmacological mechanisms remain unclear. METHOD: Anti-arthritic effects of TZG were evaluated using an adjuvant-induced arthritis (AIA) rat model. Transcriptional regulatory network analysis based on synovial tissues obtained from AIA rats, combining with our previous analysis based on whole blood samples from RA patients with cold-dampness syndrome and co-immunoprecipitation were performed to identify involved dominant pathways, which were experimentally verified using AIA-wind-cold-dampness stimulation modified (AIA-M) animal model. RESULTS: TZG treatment dramatically attenuated joint injury and inflammatory response in AIA rats, and PSMC2-RUNX2-COL1A1 axis, which was closely associated with bone/cartilage damage, was inferred to be one of therapeutic targets of TZG against RA. Experimentally, TZG displayed obvious pharmacological effects for alleviating the joint inflammation and destruction through reinstating the body weight, reducing the arthritis score, the limbs diameters, the levels of RF and CRP, and the inflammatory cytokines, recovering the thymus and spleen indexes, diminishing bone and cartilage destruction, as well elevating the pain thresholds of AIA-M rats. In addition, TZG markedly reversed the abnormal energy metabolism in AIA-M rats through enhancing articular temperature, daily water consumption, and regulating expression levels of energy metabolism parameters and hormones. Moreover, TZG also significantly modulated the abnormal expression levels of PSMC2, RUNX2 and COL1A1 proteins in the ankle tissues of AIA-M rats. CONCLUSION: TZG may exert the bone protective effects in RA therapy via regulating bone and cartilage damage-associated PSMC2-RUNX2-COL1A1 axis.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Chaine alpha-1 du collagène de type I , Collagène de type I , Médicaments issus de plantes chinoises , Animaux , Polyarthrite rhumatoïde/traitement médicamenteux , Arthrite expérimentale/traitement médicamenteux , Rats , Humains , Mâle , Médicaments issus de plantes chinoises/usage thérapeutique , Médicaments issus de plantes chinoises/pharmacologie , Collagène de type I/métabolisme , Collagène de type I/génétique , Réseaux de régulation génique/effets des médicaments et des substances chimiques , Sous-unité alpha 1 du facteur CBF/génétique , Sous-unité alpha 1 du facteur CBF/métabolisme , Cartilage/métabolisme , Cartilage/anatomopathologie , Cartilage/effets des médicaments et des substances chimiques , Os et tissu osseux/effets des médicaments et des substances chimiques , Os et tissu osseux/métabolisme , Os et tissu osseux/anatomopathologie , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques
18.
Biomolecules ; 14(7)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-39062525

RÉSUMÉ

Peptide-based drug development is a promising direction due to its excellent biological activity, minimal immunogenicity, high in vivo stability, and efficient tissue penetrability. GV1001, an amphiphilic peptide, has proven effective as an anti-cancer vaccine, but its effect on osteoblast differentiation is unknown. To identify proteins interacting with GV1001, biotin-conjugated GV1001 was constructed and confirmed by mass spectrometry. Proteomic analyses were performed to determine GV1001's interaction with osteogenic proteins. GV1001 was highly associated with peptidyl-prolyl isomerase A and co-immunoprecipitation assays revealed that GV1001 bound to peptidyl-prolyl cis-trans isomerase 1 (Pin1). GV1001 significantly increased alkaline phosphatase (ALP) activity, bone nodule formation, and the expression of osteogenic gene markers. GV1001-induced osteogenic activity was enhanced by Pin1 overexpression and abolished by Pin1 knockdown. GV1001 increased the protein stability and transcriptional activity of Runx2 and Osterix. Importantly, GV1001 administration enhanced bone mass density in the OVX mouse model, as verified by µCT analysis. GV1001 demonstrated protective effects against bone loss in OVX mice by upregulating osteogenic differentiation via the Pin1-mediated protein stabilization of Runx2 and Osterix. GV1001 could be a potential candidate with anabolic effects for the prevention and treatment of osteoporosis.


Sujet(s)
Peptides de pénétration cellulaire , Sous-unité alpha 1 du facteur CBF , NIMA-interacting peptidylprolyl isomerase , Ostéogenèse , Facteur de transcription Sp7 , Animaux , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Ostéogenèse/effets des médicaments et des substances chimiques , NIMA-interacting peptidylprolyl isomerase/métabolisme , NIMA-interacting peptidylprolyl isomerase/génétique , Souris , Peptides de pénétration cellulaire/pharmacologie , Peptides de pénétration cellulaire/composition chimique , Facteur de transcription Sp7/métabolisme , Facteur de transcription Sp7/génétique , Humains , Femelle , Stabilité protéique/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Ostéoblastes/cytologie
19.
Crit Rev Eukaryot Gene Expr ; 34(7): 51-66, 2024.
Article de Anglais | MEDLINE | ID: mdl-39072409

RÉSUMÉ

The RUNX2 transcription factor was discovered as an essential transcriptional regulator for commitment to osteoblast lineage cells and bone formation. Expression of RUNX2 in other tissues, such as breast, prostate, and lung, has been linked to oncogenesis, cancer progression, and metastasis. In this study, we sought to determine the extent of RUNX2 involvement in other tumors using a pan-cancer analysis strategy. We correlated RUNX2 expression and clinical-pathological parameters in human cancers by interrogating publicly available multiparameter clinical data. Our analysis demonstrated that altered RUNX2 expression or function is associated with several cancer types from different tissues. We identified three tumor types associated with increased RUNX2 expression and four other tumor types associated with decreased RUNX2 expression. Our pan-cancer analysis for RUNX2 revealed numerous other discoveries for RUNX2 regulation of different cancers identified in each of the pan-cancer databases. Both up and down regulation of RUNX2 was observed during progression of specific types of cancers in promoting the distinct types of cancers.


Sujet(s)
Sous-unité alpha 1 du facteur CBF , Régulation de l'expression des gènes tumoraux , Tumeurs , Humains , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Pronostic , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme
20.
Sci Rep ; 14(1): 17344, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39069521

RÉSUMÉ

To investigate the mechanism of curcumin (CUR) on vascular calcification (VC), we screen for common targets of CUR and atherosclerosis and verify the targets genes in vivo and in vitro experiments. The common targets of CUR and AS were screened and obtained using different databases. These target genes were analyzed by GO and KEGG pathway enrichment analysis. PPI network analysis was performed and to analyze the key targets. A rat VC model was constructed and CUR was fed for three weeks. The changes of vascular structure and calcium salt deposition were observed in H&E and Von Kossa staining. Further, the expression of these target proteins was detected in the primary VSMCs of VC. The 31 common targets were obtained. GO functional enrichment analysis obtained 1284 terms and KEGG pathway enriched 66 pathways. The key genes were identified in the cytoHubba plugin. The molecular docking analysis showed that CUR bound strongly to EGFR, STAT3 and BCL2. The animal experiments showed the deposition calcium salt reduced by the CUR administration. These proteins BMP2, RUNX2, EGFR, STAT3 and BAX expression were upregulated in VC group and CUR attenuated the upregulated expression. The signal protein Akt and p65 expression increased in VC group and decreased in CUR group. We identified some common target genes of CUR and AS and identified these key genes. The anti-VC effect of CUR was associated with the inhibition of upregulation of EGFR, STAT3 and RUNX2 expression in VSMCs.


Sujet(s)
Sous-unité alpha 1 du facteur CBF , Curcumine , Simulation de docking moléculaire , Facteur de transcription STAT-3 , Calcification vasculaire , Animaux , Curcumine/pharmacologie , Calcification vasculaire/métabolisme , Calcification vasculaire/traitement médicamenteux , Calcification vasculaire/anatomopathologie , Rats , Facteur de transcription STAT-3/métabolisme , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Récepteurs ErbB/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Protéine morphogénétique osseuse de type 2/métabolisme , Protéine morphogénétique osseuse de type 2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Protéine Bax/métabolisme , Protéine Bax/génétique , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE