Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.850
Filtrer
1.
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
2.
Arch Oral Biol ; 165: 106027, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38870610

RÉSUMÉ

OBJECTIVE: This study examined how range concentrations of Fibroblast Growth Factor-2 (FGF-2) influence the differentiation and activity of human-derived periodontal ligament (hPDLSCs) and alveolar bone-derived stem cells (haBMSCs). DESIGN: hPDLSCs and haBMSCs were cultured with varying concentrations of FGF-2 (0, 1, 2.5, 5, 10, 20 ng/mL) and monitored for osteogenic differentiation through alkaline phosphatase (ALP) activity and quantification of gene expression (qRT-PCR) for osteogenesis markers. Additionally, alizarin red staining and a hydroxyproline colorimetric assay evaluated and quantified osteogenic matrix mineralization and collagen deposition. Statistical analyses were performed using one-way ANOVA or two-way ANOVA for multiple comparisons between groups. RESULTS: At low FGF-2 concentrations, hPDLSCs differentiated toward an osteogenic lineage, whereas higher concentrations of FGF-2 inhibited osteogenesis and promoted fibroblastic differentiation. The effect of FGF-2 at the lowest concentration tested (1 ng/mL) led to significantly higher ALP activity than osteogenically induced positive controls at early time points and equivalent RUNX2 expression at early and later time points. FGF-2 supplementation of haBMSC cultures was sufficient, at all concentrations, to increase ALP activity at an earlier time point. Mineralization of haBMSC cultures increased significantly within 5-20 ng/mL FGF-2 concentrations under basal growth media conditions (α-minimal essential medium supplemented with 15 % fetal bovine serum and 1 % penicillin/streptomycin). CONCLUSIONS: FGF-2 has a dual capacity in promoting osteogenic and fibroblastic differentiation within hPDLSCs contingent upon the dosage and timing of administration, alongside supporting osteogenic differentiation in haBMSCs. These findings underscore the need for precision growth factors dosing when considering the design of biomaterials for periodontal regeneration.


Sujet(s)
Phosphatase alcaline , Différenciation cellulaire , Facteur de croissance fibroblastique de type 2 , Ostéogenèse , Desmodonte , Desmodonte/cytologie , Desmodonte/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Facteur de croissance fibroblastique de type 2/pharmacologie , Humains , Ostéogenèse/effets des médicaments et des substances chimiques , Ostéogenèse/physiologie , Cellules cultivées , Phosphatase alcaline/métabolisme , Processus alvéolaire/cytologie , Processus alvéolaire/effets des médicaments et des substances chimiques , Cellules souches/effets des médicaments et des substances chimiques , Sous-unité alpha 1 du facteur CBF/métabolisme , Réaction de polymérisation en chaine en temps réel
3.
Aging (Albany NY) ; 16(12): 10446-10461, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38885076

RÉSUMÉ

Ferroptosis is a new way of cell death, and stimulating the process of cell ferroptosis is a new strategy to treat breast cancer. NGR1 has good anti-cancer activity and is able to slow the progression of breast cancer. However, NGR1 has not been reported in the field related to ferroptosis. By searching the online database for potential targets of NGR1 and the breast cancer disease database, among 11 intersecting genes we focused on Runt-related transcription factor 2 (RUNX2), which is highly expressed in breast cancer, and KEGG pathway enrichment showed that the intersecting genes were mainly enriched in the AGE (advanced glycosylation end products)-RAGE (receptor of AGEs) signaling pathway. After that, we constructed overexpression and down-regulation breast cancer cell lines of RUNX2 in vitro, and tested whether NGR1 treatment induced ferroptosis in breast cancer cells by regulating RUNX2 to inhibit the AGE-RAGE signaling pathway through phenotyping experiments of ferroptosis, Western blot experiments, QPCR experiments, and electron microscopy observation. The results showed that NGR1 was able to inhibit the expression level of RUNX2 and suppress the AGE/PAGE signaling pathway in breast cancer cells. NGR1 was also able to promote the accumulation of Fe2+ and oxidative damage in breast cancer cells by regulating RUNX2 and then down-regulating the expression level of GPX4, FIH1 and up-regulating the expression level of ferroptosis-related proteins such as COX2, ACSL4, PTGS2 and NOX1, which eventually led to the ferroptosis of breast cancer cells.


Sujet(s)
Tumeurs du sein , Sous-unité alpha 1 du facteur CBF , Ferroptose , Transduction du signal , Ferroptose/effets des médicaments et des substances chimiques , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Femelle , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Ginsénosides/pharmacologie , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Récepteur spécifique des produits finaux de glycosylation avancée/génétique , Produits terminaux de glycation avancée/métabolisme , Cellules MCF-7
4.
J Orthop Surg Res ; 19(1): 330, 2024 Jun 02.
Article de Anglais | MEDLINE | ID: mdl-38825686

RÉSUMÉ

OBJECTIVE: The present study aimed to investigate the underlying mechanism of mechanical stimulation in regulating osteogenic differentiation. MATERIALS AND METHODS: Osteoblasts were exposed to compressive force (0-4 g/cm2) for 1-3 days or CGRP for 1 or 3 days. Expression of receptor activity modifying protein 1 (RAMP1), the transcription factor RUNX2, osteocalcin, p38 and p-p38 were analyzed by western blotting. Calcium mineralization was analyzed by alizarin red straining. RESULTS: Using compressive force treatments, low magnitudes (1 and 2 g/cm2) of compressive force for 24 h promoted osteoblast differentiation and mineral deposition whereas higher magnitudes (3 and 4 g/cm2) did not produce osteogenic effect. Through western blot assay, we observed that the receptor activity-modifying protein 1 (RAMP1) expression was upregulated, and p38 mitogen-activated protein kinase (MAPK) was phosphorylated during low magnitudes compressive force-promoted osteoblast differentiation. Further investigation of a calcitonin gene-related peptide (CGRP) peptide incubation, a ligand for RAMP1, showed that CGRP at concentration of 25 and 50 ng/ml could increase expression levels of RUNX2 and osteocalcin, and percentage of mineralization, suggesting its osteogenic potential. In addition, with the same conditions, CGRP also significantly upregulated RAMP1 and phosphorylated p38 expression levels. Also, the combination of compressive forces (1 and 2 g/cm2) with 50 ng/ml CGRP trended to increase RAMP1 expression, p38 activity, and osteogenic marker RUNX2 levels, as well as percentage of mineralization compared to compressive force alone. This suggest that RAMP1 possibly acts as an upstream regulator of p38 signaling during osteogenic differentiation. CONCLUSION: These findings suggest that CGRP-RAMP1/p38MAPK signaling implicates in osteoblast differentiation in response to optimal magnitude of compressive force. This study helps to define the underlying mechanism of compressive stimulation and may also enhance the application of compressive stimulation or CGRP peptide as an alternative approach for accelerating tooth movement in orthodontic treatment.


Sujet(s)
Différenciation cellulaire , Ostéoblastes , Ostéogenèse , Protéine-1 modifiant l'activité des récepteurs , p38 Mitogen-Activated Protein Kinases , Ostéoblastes/physiologie , Ostéoblastes/métabolisme , Ostéoblastes/cytologie , Différenciation cellulaire/physiologie , Protéine-1 modifiant l'activité des récepteurs/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme , Ostéogenèse/physiologie , Peptide relié au gène de la calcitonine/métabolisme , Système de signalisation des MAP kinases/physiologie , Contrainte mécanique , Animaux , Cellules cultivées , Sous-unité alpha 1 du facteur CBF/métabolisme , Transduction du signal/physiologie , Ostéocalcine/métabolisme
5.
Mol Biol Rep ; 51(1): 702, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38822942

RÉSUMÉ

BACKGROUND: The development of cost-effective, simple, environment-friendly biographene is an area of interest. To accomplish environmentally safe, benign culturing that has advantages over other methods to reduce the graphene oxide (GO), extracellular metabolites from actinobacteria associated with mushrooms were used for the first time. METHODS: Bactericidal effect of GO against methicillin-resistant Staphylococcus aureus, antioxidant activity, and hydroxyapatite-like bone layer formation, gene expression analysis and appropriate biodegradation of the microbe-mediated synthesis of graphene was studied. RESULTS: Isolated extracellular contents Streptomyces achromogenes sub sp rubradiris reduced nano-GO to graphene (rGO), which was further examined by spectrometry and suggested an efficient conversion and significant reduction in the intensity of all oxygen-containing moieties and shifted crystalline peaks. Electron microscopic results also suggested the reduction of GO layer. In addition, absence of significant toxicity in MG-63 cell line, intentional free radical scavenging prowess, liver and kidney histopathology, and Wistar rat bone regeneration through modulation of OPG/RANKL/RUNX2/ALP pathways show the feasibility of the prepared nano GO. CONCLUSIONS: The study demonstrates the successful synthesis of biographene from actinobacterial extracellular metabolites, its potential biomedical applications, and its promising role in addressing health and environmental concerns.


Sujet(s)
Régénération osseuse , Graphite , Ostéoprotégérine , Ligand de RANK , Rat Wistar , Graphite/pharmacologie , Animaux , Régénération osseuse/effets des médicaments et des substances chimiques , Rats , Ligand de RANK/métabolisme , Ostéoprotégérine/métabolisme , Humains , Matériaux biocompatibles/pharmacologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Actinobacteria/métabolisme , Antibactériens/pharmacologie , Antioxydants/métabolisme , Antioxydants/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques
6.
Sci Rep ; 14(1): 13441, 2024 06 11.
Article de Anglais | MEDLINE | ID: mdl-38862780

RÉSUMÉ

The present study aims to explore the etiology of Diabetic osteoporosis (DOP), a chronic complication associated with diabetes mellitus. Specifically, the research seeks to identify potential miRNA biomarkers of DOP and investigated role in regulating osteoblasts. To achieve this, an animal model of DOP was established through the administration of a high-sugar and high-fat diet, and then injection of streptozotocin. Bone microarchitecture and histopathology analysis were analyzed. Rat calvarial osteoblasts (ROBs) were stimulated with high glucose (HG). MiRNA profiles of the stimulated osteoblasts were compared to control osteoblasts using sequencing. Proliferation and mineralization abilities were assessed using MTT assay, alkaline phosphatase, and alizarin red staining. Expression levels of OGN, Runx2, and ALP were determined through qRT-PCR and Western blot. MiRNA-sequencing results revealed increased miRNA-702-5p levels. Luciferase reporter gene was utilized to study the correlation between miR-702-5p and OGN. High glucose impaired cell proliferation and mineralization in vitro by inhibiting OGN, Runx2, and ALP expressions. Interference with miR-702-5p decreased OGN, Runx2, and ALP levels, which were restored by OGN overexpression. Additionally, downregulation of OGN and Runx2 in DOP rat femurs was confirmed. Therefore, the miRNA-702-5p/OGN/Runx2 signaling axis may play a role in DOP, and could be diagnostic biomarker and therapeutic target for not only DOP but also other forms of osteoporosis.


Sujet(s)
Glucose , microARN , Ostéoblastes , Ostéoporose , Animaux , microARN/génétique , microARN/métabolisme , Ostéoblastes/métabolisme , Ostéoporose/génétique , Ostéoporose/métabolisme , Ostéoporose/anatomopathologie , Ostéoporose/étiologie , Rats , Glucose/métabolisme , Glucose/pharmacologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Prolifération cellulaire , Diabète expérimental/métabolisme , Diabète expérimental/génétique , Mâle , Rat Sprague-Dawley
7.
Sci Rep ; 14(1): 12704, 2024 06 03.
Article de Anglais | MEDLINE | ID: mdl-38830996

RÉSUMÉ

To explore the role of YAP, a key effector of the Hippo pathway, in temporomandibular joint (TMJ) ankylosis. The temporal and spatial expression of YAP was detected via immunohistochemistry and multiplex immunohistochemistry on postoperative Days 1, 4, 7, 9, 11, 14 and 28 in a sheep model. Isolated mesenchymal stem cells (MSCs) from samples of the Day 14. The relative mRNA expression of YAP was examined before and after the osteogenic induction of MSCs. A YAP-silenced MSC model was constructed, and the effect of YAP knockdown on MSC function was examined. YAP is expressed in the nucleus of the key sites that determine the ankylosis formation, indicating that YAP is activated in a physiological state. The expression of YAP increased gradually over time. Moreover, the number of cells coexpressing of RUNX2 and YAP-with the osteogenic active zone labelled by RUNX2-tended to increase after Day 9. After the osteogenic induction of MSCs, the expression of YAP increased. After silencing YAP, the osteogenic, proliferative and migratory abilities of the MSCs were inhibited. YAP is involved in the early development of TMJ bony ankylosis. Inhibition of YAP using shRNA might be a promising way to prevent or treat TMJ ankylosis.


Sujet(s)
Ankylose , Cellules souches mésenchymateuses , Ostéogenèse , Troubles de l'articulation temporomandibulaire , Animaux , Cellules souches mésenchymateuses/métabolisme , Troubles de l'articulation temporomandibulaire/métabolisme , Troubles de l'articulation temporomandibulaire/anatomopathologie , Troubles de l'articulation temporomandibulaire/génétique , Ankylose/métabolisme , Ankylose/anatomopathologie , Ankylose/génétique , Protéines de signalisation YAP/métabolisme , Articulation temporomandibulaire/métabolisme , Articulation temporomandibulaire/anatomopathologie , Ovis , Prolifération cellulaire , Modèles animaux de maladie humaine , Différenciation cellulaire , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Mouvement cellulaire , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique
8.
Injury ; 55(7): 111627, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38834011

RÉSUMÉ

BACKGROUND: As a two-stage surgical procedure, Masquelet's technique has been used to care for critical-size bone defects (CSD). We aimed to determine the effects of modified and altered bone cement with biological or chemical enriching agents on the progression of Masquelet's induced membrane (IM) applied to a rat femur CSD model, and to compare the histopathological, biochemical, and immunohistochemical findings of these cements to enhance IM capacity. METHODS: Thirty-five male rats were included in five groups: plain polymethyl methacrylate (PMMA), estrogen-impregnated PMMA (E+PMMA), bone chip added PMMA (BC+PMMA), hydroxyapatite-coated PMMA (HA) and calcium phosphate cement (CPC). The levels of bone alkaline phosphatase (BALP), osteocalcin (OC), and tumor necrosis factor-alpha (TNF-α) were analyzed in intracardiac blood samples collected at the end of 4 weeks of the right femur CSD intervention. All IMs collected were fixed and prepared for histopathological scoring. The tissue levels of rat-specific Transforming Growth Factor-Beta (TGF-ß), Runt-related Transcription Factor 2 (Runx2), and Vascular Endothelial Growth Factor (VEGF) were analyzed immunohistochemically. RESULTS: Serum levels of BALP and OC were significantly higher in E+PMMA and BC+PMMA groups than those of other groups (P = 0.0061 and 0.0019, respectively). In contrast, TNF-α levels of all groups with alternative bone cement significantly decreased compared to bare PMMA (P = 0.0116). Histopathological scores of E+PMMA, BC+PMMA, and CPC groups were 6.86 ± 1.57, 4.71 ± 0.76, and 6.57 ± 1.51, respectively, which were considerably higher than those of PMMA and HA groups (3.14 ± 0.70 and 1.86 ± 0.69, respectively) (P < 0.0001). Significant increases in TGF-ß and VEGF expressions were observed in E+PMMA and CPC groups (P = 0.0001 and <0.0001, respectively) whereas Runx2 expression significantly increased only in the HA group compared to other groups (P < 0.0001). CONCLUSIONS: The modified PMMA with E and BC, and CPC as an alternative spacer resulted in a well-differentiated IM and increased IM progression by elevating BALP and OC levels in serum and by mediating expressions of TGF-ß and VEGF at the tissue level. Estrogen-supplemented cement spacer has yielded promising findings between modified and alternative bone cement.


Sujet(s)
Ciments osseux , Modèles animaux de maladie humaine , Fémur , Poly(méthacrylate de méthyle) , Facteur de croissance endothéliale vasculaire de type A , Animaux , Rats , Mâle , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Fémur/anatomopathologie , Fémur/effets des médicaments et des substances chimiques , Fractures du fémur/anatomopathologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Ostéocalcine/métabolisme , Phosphatase alcaline/métabolisme , Facteur de croissance transformant bêta/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Rat Sprague-Dawley , Phosphates de calcium , Consolidation de fracture/effets des médicaments et des substances chimiques , Consolidation de fracture/physiologie , Régénération osseuse/effets des médicaments et des substances chimiques , Durapatite
9.
Genome Res ; 34(5): 680-695, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38777607

RÉSUMÉ

Gastric cancer (GC) is the fifth most common cancer worldwide and is a heterogeneous disease. Among GC subtypes, the mesenchymal phenotype (Mes-like) is more invasive than the epithelial phenotype (Epi-like). Although gene expression of the epithelial-to-mesenchymal transition (EMT) has been studied, the regulatory landscape shaping this process is not fully understood. Here we use ATAC-seq and RNA-seq data from a compendium of GC cell lines and primary tumors to detect drivers of regulatory state changes and their transcriptional responses. Using the ATAC-seq data, we developed a machine learning approach to determine the transcription factors (TFs) regulating the subtypes of GC. We identified TFs driving the mesenchymal (RUNX2, ZEB1, SNAI2, AP-1 dimer) and the epithelial (GATA4, GATA6, KLF5, HNF4A, FOXA2, GRHL2) states in GC. We identified DNA copy number alterations associated with dysregulation of these TFs, specifically deletion of GATA4 and amplification of MAPK9 Comparisons with bulk and single-cell RNA-seq data sets identified activation toward fibroblast-like epigenomic and expression signatures in Mes-like GC. The activation of this mesenchymal fibrotic program is associated with differentially accessible DNA cis-regulatory elements flanking upregulated mesenchymal genes. These findings establish a map of TF activity in GC and highlight the role of copy number driven alterations in shaping epigenomic regulatory programs as potential drivers of GC heterogeneity and progression.


Sujet(s)
Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , Apprentissage machine , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Transition épithélio-mésenchymateuse/génétique , Facteur de transcription AP-1/métabolisme , Facteur de transcription AP-1/génétique , Lignée cellulaire tumorale , Fibrose/génétique , Sous-unité alpha 1 du facteur CBF/génétique , Sous-unité alpha 1 du facteur CBF/métabolisme , Variations de nombre de copies de segment d'ADN , Sous-unité alpha 2 du facteur CBF
10.
J Bone Miner Metab ; 42(3): 290-301, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38796648

RÉSUMÉ

INTRODUCTION: Osteoporosis is a significant health concern characterized by weak and porous bones, particularly affecting menopausal women aged 50 and above, leading to increased risk of hip fractures and associated morbidity and mortality. MATERIALS AND METHODS: We conducted a study to assess the efficacy of single-strain versus mixed-strain probiotic supplementation on bone health using an ovariectomy (OVX) rat model of induced bone loss. The probiotics evaluated were Lactobacillus helveticus (L. helveticus), Bifidobacterium longum (B. longum), and a combination of both. Rats were divided into five groups: SHAM (Control negative), OVX (Control positive), OVX +L. helveticus, OVX + B. longum, and OVX + mixed L. helveticus and B. longum. Daily oral administration of probiotics at 10^8-10^9 CFU/mL began two weeks post-surgery and continued for 16 weeks. RESULTS: Both single-strain and mixed-strain probiotic supplementation upregulated expression of osteoblastic genes (BMP- 2, RUNX-2, OSX), increased serum osteocalcin (OC) levels, and improved bone formation parameters. Serum C-terminal telopeptide (CTX) levels and bone resorption parameters were reduced. However, the single-strain supplementation demonstrated superior efficacy compared to the mixed-strain approach. CONCLUSION: Supplementation with B. longum and L. helveticus significantly reduces bone resorption and improves bone health in OVX rats, with single-strain supplementation showing greater efficacy compared to a mixed-strain combination. These findings highlight the potential of probiotics as a therapeutic intervention for osteoporosis, warranting further investigation in human studies.


Sujet(s)
Densité osseuse , Fémur , Lactobacillus helveticus , Ostéoblastes , Ovariectomie , Probiotiques , ARN messager , Animaux , Probiotiques/pharmacologie , Probiotiques/administration et posologie , Femelle , Rats , Ostéoblastes/métabolisme , Fémur/métabolisme , ARN messager/métabolisme , ARN messager/génétique , Compléments alimentaires , Rat Sprague-Dawley , Bifidobacterium longum , Ostéoporose/métabolisme , Ostéocalcine/sang , Ostéocalcine/métabolisme , Régulation de l'expression des gènes , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique
11.
Biochem Biophys Res Commun ; 722: 150155, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-38795454

RÉSUMÉ

Runt-related transcription factor (RUNX) family members play critical roles in the development of multiple organs. Mammalian RUNX family members, consisting of RUNX1, RUNX2, and RUNX3, have distinct tissue-specific expression and function. In this study, we examined the spatiotemporal expression patterns of RUNX family members in developing kidneys and analyzed the role of RUNX1 during kidney development. In the developing mouse kidney, RUNX1 protein was strongly expressed in the ureteric bud (UB) tip and weakly expressed in the distal segment of the renal vesicle (RV), comma-shaped body (CSB), and S-shaped body (SSB). In contrast, RUNX2 protein was restricted to the stroma, and RUNX3 protein was only expressed in immune cells. We also analyzed the expression of RUNX family members in the cynomolgus monkey kidney. We found that expression patterns of RUNX2 and RUNX3 were conserved between rodents and primates, whereas RUNX1 was only expressed in the UB tip, not in the RV, CSB, or SSB of cynomolgus monkeys, suggesting a species differences. We further evaluated the roles of RUNX1 using two different conditional knockout mice: Runx1f/f:HoxB7-Cre and Runx1f/f:R26-CreERT2 and found no abnormalities in the kidney. Our findings showed that RUNX1, which is mainly expressed in the UB tip, is not essential for kidney development.


Sujet(s)
Sous-unité alpha 2 du facteur CBF , Rein , Animaux , Sous-unité alpha 2 du facteur CBF/métabolisme , Sous-unité alpha 2 du facteur CBF/génétique , Rein/métabolisme , Rein/embryologie , Rein/croissance et développement , Souris , Macaca fascicularis , Régulation de l'expression des gènes au cours du développement , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Sous-unité alpha 3 du facteur CBF/métabolisme , Sous-unité alpha 3 du facteur CBF/génétique , Sous-unités alpha du facteur CBF/métabolisme , Sous-unités alpha du facteur CBF/génétique , Souris de lignée C57BL , Souris knockout
12.
Redox Biol ; 73: 103183, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38759418

RÉSUMÉ

AIMS: Vascular calcification is strongly linked to the development of major adverse cardiovascular events, but effective treatments are lacking. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are an emerging category of oral hypoglycemic drugs that have displayed marked effects on metabolic and cardiovascular diseases, including recently reported vascular medial calcification. However, the roles and underlying mechanisms of SGLT2 inhibitors in vascular calcification have not been fully elucidated. Thus, we aimed to further determine whether SGLT2 inhibitors protect against vascular calcification and to investigate the mechanisms involved. METHODS AND RESULTS: A computed tomography angiography investigation of coronary arteries from 1554 patients with type 2 diabetes revealed that SGLT2 inhibitor use was correlated with a lower Agatston calcification score. In the vitamin D3 overdose, 5/6 nephrectomy chronic kidney disease-induced medial calcification and Western diet-induced atherosclerotic intimal calcification models, dapagliflozin (DAPA) substantially alleviated vascular calcification in the aorta. Furthermore, we showed that DAPA reduced vascular calcification via Runx2-dependent osteogenic transdifferentiation in vascular smooth muscle cells (VSMCs). Transcriptome profiling revealed that thioredoxin domain containing 5 (TXNDC5) was involved in the attenuation of vascular calcification by DAPA. Rescue experiments showed that DAPA-induced TXNDC5 downregulation in VSMCs blocked the protective effect on vascular calcification. Furthermore, TXNDC5 downregulation disrupted protein folding-dependent Runx2 stability and promoted subsequent proteasomal degradation. Moreover, DAPA downregulated TXNDC5 expression via amelioration of oxidative stress and ATF6-dependent endoplasmic reticulum stress. Consistently, the class effects of SGLT2 inhibitors on vascular calcification were validated with empagliflozin in intimal and medial calcification models. CONCLUSIONS: SGLT2 inhibitors ameliorate vascular calcification through blocking endoplasmic reticulum stress-dependent TXNDC5 upregulation and promoting subsequent Runx2 proteasomal degradation, suggesting that SGLT2 inhibitors are potentially beneficial for vascular calcification treatment and prevention.


Sujet(s)
Glucosides , Ostéogenèse , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Calcification vasculaire , Calcification vasculaire/métabolisme , Calcification vasculaire/traitement médicamenteux , Calcification vasculaire/anatomopathologie , Calcification vasculaire/étiologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Animaux , Humains , Ostéogenèse/effets des médicaments et des substances chimiques , Souris , Glucosides/pharmacologie , Mâle , Thiorédoxines/métabolisme , Thiorédoxines/génétique , Composés benzhydryliques/pharmacologie , Diabète de type 2/métabolisme , Diabète de type 2/complications , Diabète de type 2/traitement médicamenteux , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/effets des médicaments et des substances chimiques , Rats , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Modèles animaux de maladie humaine , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/cytologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Femelle
13.
Article de Anglais | MEDLINE | ID: mdl-38788347

RÉSUMÉ

BACKGROUND: Linoleic acid (LNA), an essential polyunsaturated fatty acid (PUFA), plays a crucial role in cellular functions. However, excessive intake of LNA, characteristic of Western diets, can have detrimental effects on cells and organs. Human observational studies have shown an inverse relationship between plasma LNA concentrations and bone mineral density. The mechanism by which LNA impairs the skeleton is unclear, and there is a paucity of research on the effects of LNA on bone-forming osteoblasts. METHODS: The effect of LNA on osteoblast differentiation, cellular bioenergetics, and production of oxidized PUFA metabolites in vitro, was studied using primary mouse bone marrow stromal cells (BMSC) and MC3T3-E1 osteoblast precursors. RESULTS: LNA treatment decreased alkaline phosphatase activity, an early marker of osteoblast differentiation, but had no effect on committed osteoblasts or on mineralization by differentiated osteoblasts. LNA suppressed osteoblast commitment by blunting the expression of Runx2 and Osterix, key transcription factors involved in osteoblast differentiation, and other key osteoblast-related factors involved in bone formation. LNA treatment was associated with increased production of oxidized LNA- and arachidonic acid-derived metabolites and blunted oxidative phosphorylation, resulting in decreased ATP production. CONCLUSION: Our results show that LNA inhibited early differentiation of osteoblasts and this inhibitory effect was associated with increased production of oxidized PUFA metabolites that likely impaired energy production via oxidative phosphorylation.


Sujet(s)
Différenciation cellulaire , Acide linoléique , Ostéoblastes , Phosphorylation oxydative , Animaux , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Ostéoblastes/cytologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Souris , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Acide linoléique/pharmacologie , Acide linoléique/métabolisme , Phosphatase alcaline/métabolisme , Sous-unité alpha 1 du facteur CBF/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Cellules cultivées
14.
Mol Biol Cell ; 35(7): ar91, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38758658

RÉSUMÉ

Mechanical cues from the tissue microenvironment, such as the stiffness of the extracellular matrix, modulate cellular forms and functions. As numerous studies have shown, this modulation depends on the stiffness-dependent remodeling of cytoskeletal elements. In contrast, very little is known about how the intracellular organelles such as mitochondria respond to matrix stiffness and whether their form, function, and localization change accordingly. Here, we performed an extensive quantitative characterization of mitochondrial morphology, subcellular localization, dynamics, and membrane tension on soft and stiff matrices. This characterization revealed that while matrix stiffness affected all these aspects, matrix stiffening most distinctively led to an increased perinuclear clustering of mitochondria. Subsequently, we could identify the matrix stiffness-sensitive perinuclear localization of filamin as the key factor dictating this perinuclear clustering. The perinuclear and peripheral mitochondrial populations differed in their motility on soft matrix but surprisingly they did not show any difference on stiff matrix. Finally, perinuclear mitochondrial clustering appeared to be crucial for the nuclear localization of RUNX2 and hence for priming human mesenchymal stem cells towards osteogenesis on a stiff matrix. Taken together, we elucidate a dependence of mitochondrial localization on matrix stiffness, which possibly enables a cell to adapt to its microenvironment.


Sujet(s)
Matrice extracellulaire , Cellules souches mésenchymateuses , Mitochondries , Humains , Matrice extracellulaire/métabolisme , Mitochondries/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cytosquelette/métabolisme , Filamines/métabolisme , Sous-unité alpha 1 du facteur CBF/métabolisme , Noyau de la cellule/métabolisme , Ostéogenèse/physiologie , Différenciation cellulaire/physiologie
15.
Int J Mol Sci ; 25(10)2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38791330

RÉSUMÉ

Bone mechanotransduction is a critical process during skeletal development in embryogenesis and organogenesis. At the same time, the type and level of mechanical loading regulates bone remodeling throughout the adult life. The aberrant mechanosensing of bone cells has been implicated in the development and progression of bone loss disorders, but also in the bone-specific aspect of other clinical entities, such as the tumorigenesis of solid organs. Novel treatment options have come into sight that exploit the mechanosensitivity of osteoblasts, osteocytes, and chondrocytes to achieve efficient bone regeneration. In this regard, runt-related transcription factor 2 (Runx2) has emerged as a chief skeletal-specific molecule of differentiation, which is prominent to induction by mechanical stimuli. Polycystins represent a family of mechanosensitive proteins that interact with Runx2 in mechano-induced signaling cascades and foster the regulation of alternative effectors of mechanotransuction. In the present narrative review, we employed a PubMed search to extract the literature concerning Runx2, polycystins, and their association from 2000 to March 2024. The keywords stated below were used for the article search. We discuss recent advances regarding the implication of Runx2 and polycystins in bone remodeling and regeneration and elaborate on the targeting strategies that may potentially be applied for the treatment of patients with bone loss diseases.


Sujet(s)
Sous-unité alpha 1 du facteur CBF , Mécanotransduction cellulaire , Canaux cationiques TRPP , Humains , Sous-unité alpha 1 du facteur CBF/métabolisme , Canaux cationiques TRPP/métabolisme , Canaux cationiques TRPP/génétique , Animaux , Os et tissu osseux/métabolisme , Remodelage osseux , Régénération osseuse , Ostéocytes/métabolisme
16.
Redox Biol ; 73: 103215, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38810422

RÉSUMÉ

The prevalence of calcific aortic valve disease (CAVD) remains substantial while there is currently no medical therapy available. Forkhead box O1 (FOXO1) is known to be involved in the pathogenesis of cardiovascular diseases, including vascular calcification and atherosclerosis; however, its specific role in calcific aortic valve disease remains to be elucidated. In this study, we identified FOXO1 significantly down-regulated in the aortic valve interstitial cells (VICs) of calcified aortic valves by investigating clinical specimens and GEO database analysis. FOXO1 silencing or inhibition promoted VICs osteogenic differentiation in vitro and aortic valve calcification in Apoe-/- mice, respectively. We identified that FOXO1 facilitated the ubiquitination and degradation of RUNX2, which process was mainly mediated by SMAD-specific E3 ubiquitin ligase 2 (SMURF2). Our discoveries unveil a heretofore unacknowledged mechanism involving the FOXO1/SMURF2/RUNX2 axis in CAVD, thereby proposing the potential therapeutic utility of FOXO1 or SMURF2 as viable strategies to impede the progression of CAVD.


Sujet(s)
Sténose aortique , Valve aortique , Calcinose , Sous-unité alpha 1 du facteur CBF , Protéine O1 à motif en tête de fourche , Ubiquitin-protein ligases , Ubiquitination , Protéine O1 à motif en tête de fourche/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Animaux , Valve aortique/métabolisme , Valve aortique/anatomopathologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Souris , Humains , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Calcinose/métabolisme , Calcinose/anatomopathologie , Calcinose/génétique , Sténose aortique/métabolisme , Sténose aortique/anatomopathologie , Sténose aortique/génétique , Mâle , Ostéogenèse/génétique , Modèles animaux de maladie humaine , Différenciation cellulaire
17.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(6): 578-585, 2024 Jun 09.
Article de Chinois | MEDLINE | ID: mdl-38808418

RÉSUMÉ

Objective: To investigate the biocompatibility of coral-like barium titanate nano-piezoelectric coatings and the influence of ultrasound-excited piezoelectric effect on the early osteogenic differentiation. Methods: The barium titanate nano-piezoelectric coating (the coating group) was prepared on the surface of titanium metal by anodic oxidation, hydrothermal reaction and high-temperature annealing, and polished titanium specimens were used as control group. The surface morphology, composition, and crystal phase and hydrophilicity of the two groups of titanium specimens were characterized using scanning electron microscopy, X-ray photoelectron spectroscopy, Raman spectroscopy and contact angle meter. The piezoelectric properties of the materials were characterized by piezoresponse force microscopy. Rat bone marrow mesenchymal stem cells (BMSC) were cultured and identified and seeded the surface of titanium specimens in two groups. The cells seeded on blank culture plates were used as blank group. After low intensity pulsed ultrasound intervention, cell proliferation and live/dead staining were detected to evaluate cytocompatibility of the coatings. Alkaline phosphatase (ALP) activity of each group was detected by ALP staining kit, and the expression of osteogenesis-related genes [integrin, bone morphogenetic protein 2 (BMP-2), Runt-related transcription factor 2 (RUNX2)] was detected by real-time fluorescent quantitative PCR (RT-qPCR) to evaluate the effect of the coating on promoting the early osteogenic differentiation of BMSC. Results: The surface of titanium specimens in the coating group showed a uniform coral-like morphology, and the diameter of the coral tentacles was 70-100 nm. The main component was tetragonal barium titanate. The surface hydrophilicity of the coating group (water contact angle 10.12°± 0.93°) was significantly better than that of the control group (water contact angle 78.32°±0.71°) (F= 10 165.91, P<0.001). The coating has a stable piezoelectric property with a piezoelectric constant of about 5 pC/N. Cell experiments showed that, with or without ultrasound, the cell proliferation activity of the coating group was significantly lower than that of the blank group and the control group on the third day (P<0.05). On the fifth day, with or without ultrasound, there was no significant difference in cell proliferation activity between the three groups (P>0.05). After 7 days of culture, the ALP activity of the coating group was significantly higher than that of the blank group and the control group (P<0.05). The results of RT-qPCR showed that the mRNA expression of integrin and BMP-2 in the coating group with ultrasound was significantly higher than that in the other groups with ultrasound, and was higher than that of the coating group without ultrasound (P<0.05). The expression of integrin mRNA in the control group with ultrasound was significantly higher than that in the control group without ultrasound (P<0.05). The expression of RUNX2 mRNA in the coating group with ultrasound was significantly higher than that in the coating group without ultrasound (P<0.05). Conclusions: The coral-like barium titanate nano-piezoelectric coating exhibits favorable biocompatibility and stable piezoelectric property, and facilitates the early osteogenic differentiation of BMSC under the excitation of low-intensity pulsed ultrasound.


Sujet(s)
Composés du baryum , Différenciation cellulaire , Cellules souches mésenchymateuses , Ostéogenèse , Titane , Animaux , Cellules souches mésenchymateuses/cytologie , Rats , Matériaux revêtus, biocompatibles , Prolifération cellulaire , Cellules de la moelle osseuse/cytologie , Propriétés de surface , Protéine morphogénétique osseuse de type 2/métabolisme , Phosphatase alcaline/métabolisme , Sous-unité alpha 1 du facteur CBF/métabolisme , Anthozoa
18.
Arch Oral Biol ; 163: 105963, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38608563

RÉSUMÉ

OBJECTIVES: Orthodontic tooth movement is a mechanobiological reaction induced by appropriate forces, including bone remodeling. The mechanosensitive Piezo channels have been shown to contribute to bone remodeling. However, information about the pathways through which Piezo channels affects osteoblasts remains limited. Thus, we aimed to investigate the influence of Piezo1 on the osteogenic and osteoclast factors in osteoblasts under mechanical load. MATERIALS AND METHODS: Cyclic stretch (CS) experiments on MC3T3-E1 were conducted using a BioDynamic mechanical stretching device. The Piezo1 channel blocker GsMTx4 and the Piezo1 channel agonist Yoda1 were used 12 h before the application of CS. MC3T3-E1 cells were then subjected to 15% CS, and the expression of Piezo1, Piezo2, BMP-2, OCN, Runx2, RANKL, p-p65/p65, and ALP was measured using quantitative real-time polymerase chain reaction, western blot, alkaline phosphatase staining, and immunofluorescence staining. RESULTS: CS of 15% induced the highest expression of Piezo channel and osteoblast factors. Yoda1 significantly increased the CS-upregulated expression of Piezo1 and ALP activity but not Piezo2 and RANKL. GsMTx4 downregulated the CS-upregulated expression of Piezo1, Piezo2, Runx2, OCN, p-65/65, and ALP activity but could not completely reduce CS-upregulated BMP-2. CONCLUSIONS: The appropriate force is more suitable for promoting osteogenic differentiation in MC3T3-E1. The Piezo1 channel participates in osteogenic differentiation of osteoblasts through its influence on the expression of osteogenic factors like BMP-2, Runx2, and OCN and is involved in regulating osteoclasts by influencing phosphorylated p65. These results provide a foundation for further exploration of osteoblast function in orthodontic tooth movement.


Sujet(s)
Protéine morphogénétique osseuse de type 2 , Sous-unité alpha 1 du facteur CBF , Canaux ioniques , Ostéoblastes , Ostéogenèse , Ostéoblastes/métabolisme , Canaux ioniques/métabolisme , Animaux , Souris , Protéine morphogénétique osseuse de type 2/métabolisme , Ostéogenèse/physiologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Ostéoclastes/métabolisme , Réaction de polymérisation en chaine en temps réel , Ligand de RANK/métabolisme , Technique de Western , Contrainte mécanique , Différenciation cellulaire , Ostéocalcine/métabolisme , Phosphatase alcaline/métabolisme , Oligopeptides/pharmacologie , Mouvement dentaire , Mécanotransduction cellulaire/physiologie , Lignée cellulaire , Remodelage osseux/physiologie , Pyrazines , Venins d'araignée , Thiadiazoles , Protéines et peptides de signalisation intercellulaire
19.
Phytomedicine ; 129: 155587, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38608598

RÉSUMÉ

BACKGROUND: Osteoporosis is a prevalent metabolic bone disease in older adults. Peroxisome proliferator-activated receptor ß (PPARß), the most abundant PPAR isotype expressed in bone tissues, plays a critical role in regulating the energy metabolism of osteoblasts. However, the botanical compounds targeting PPARß for the treatment of osteoporosis remain largely unexplored. PURPOSE: To discover a potent PPARß agonist from botanical compounds, as well as to investigate the anti-osteoporosis effects and to elucidate the underlying mechanisms of the newly identified PPARß agonist. METHODS: The PPARß agonist effects of botanical compounds were screened by an in vitro luciferase reporter gene assay. The PPARß agonist effects of pectolinarigenin (PEC) in bone marrow mesenchymal stromal cells (BMSCs) were validated by Western blotting. RNA-seq transcriptome analyses were conducted to reveal the underlying osteoporosis mechanisms of PEC in BMSCs. The PPARß antagonist (GSK0660) and Wnt signaling inhibitor (XAV969) were used to explore the role of the PPARß and Wnt signaling cascade in the anti-osteoporosis effects of PEC. PEC or the PEG-PLGA nanoparticles of PEC (PEC-NP) were intraperitoneally administrated in both wild-type mice and ovariectomy-induced osteoporosis mice to examine its anti-osteoporotic effects in vivo. RESULTS: PEC, a newly identified naturally occurring PPARß agonist, significantly promotes osteogenic differentiation and up-regulates the osteogenic differentiation-related genes (Runx2, Osterix, and Bmp2) in BMSCs. RNA sequencing and functional gene enrichment analysis suggested that PEC could activate osteogenic-related signaling pathways, including Wnt and PPAR signaling pathways. Further investigations suggested that PEC could enhance Wnt/ß-catenin signaling in a PPARß-dependent manner in BMSCs. Animal tests showed that PEC-NP promoted bone mass and density, increased the bone cell matrix protein, and accelerated bone formation in wild-type mice, while PEC-NP also played a preventive role in ovariectomy-induced osteoporosis mice via maintaining the expression level of bone cell matrix protein, balancing the rate of bone formation, and slowing down bone loss. Additionally, PEC-NP did not cause any organ injury and body weight loss after long-term use (11 weeks). CONCLUSION: PEC significantly promotes bone formation and reduces bone loss in both BMSCs and ovariectomy-induced osteoporosis mice via enhancing the Wnt signaling cascade in a PPARß-dependent manner, providing a new alternative therapy for preventing estrogen deficiency-induced osteoporotic diseases.


Sujet(s)
Cellules souches mésenchymateuses , Souris de lignée C57BL , Ostéoporose , Récepteur PPAR bêta , Voie de signalisation Wnt , Animaux , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Ostéoporose/traitement médicamenteux , Récepteur PPAR bêta/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Femelle , Souris , Ostéogenèse/effets des médicaments et des substances chimiques , Ovariectomie , Saponines/pharmacologie , Protéine morphogénétique osseuse de type 2/métabolisme , Sous-unité alpha 1 du facteur CBF/métabolisme , 4H-1-Benzopyran-4-ones , Sulfones , Thiophènes
20.
J Orthop Surg Res ; 19(1): 260, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38659042

RÉSUMÉ

Circular RNAs (circRNAs) are a newly appreciated type of endogenous noncoding RNAs that play vital roles in the development of various human cancers, including osteosarcoma (OS). In this study, we investigated three circRNAs (circ_0076684, circ_0003563, circ_0076691) from the RUNX Family Transcription Factor 2 (RUNX2) gene locus in OS. We found that the expression of circ_0076684, circ_0003563, circ_0076691, and RUNX2 mRNA is upregulated in OS, which is a consequence of CBX4-mediated transcriptional activation. Among these three RUNX2-circRNAs, only circ_0076684 is significantly associated with the clinical features and prognosis of OS patients. Functional experiments indicate that circ_0076684 promotes OS progression in vitro and in vivo. Circ_0076684 acts as a sponge for miR-370-3p, miR-140-3p, and miR-193a-5p, raising Cut Like Homeobox 1 (CUX1) expression by sponging these three miRNAs. Furthermore, we presented that circ_0076684 facilitates OS progression via CUX1. In conclusion, this study found that the expression of three circRNAs and RUNX2 mRNA from the RUNX2 gene locus is significantly upregulated in OS, as a result of CBX4-mediated transcriptional activation. Circ_0076684 raises CUX1 expression by sponging miR-370-3p, miR-140-3p, and miR-193a-5p, and facilitates OS progression via CUX1.


Sujet(s)
Tumeurs osseuses , Sous-unité alpha 1 du facteur CBF , Ligases , microARN , Ostéosarcome , Protéines du groupe Polycomb , ARN circulaire , Régulation positive , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , Humains , ARN circulaire/génétique , microARN/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Sous-unité alpha 1 du facteur CBF/métabolisme , Régulation de l'expression des gènes tumoraux/génétique , Mâle , Animaux , Évolution de la maladie , Lignée cellulaire tumorale , Femelle , Activation de la transcription/génétique , Pronostic , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Souris , Protéines de répression/génétique , Protéines de répression/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...