Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.516
Filtrer
1.
J Orofac Orthop ; 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093345

RÉSUMÉ

AIMS: To investigate the effect of tumor necrosis factor (TNF) on the growth of human periodontal ligament (PDL) cells, their osteogenic differentiation and modulation of their matrix secretion in vitro. METHODS: The influence of 10 ng/ml TNF on proliferation and metabolic activity of PDL cells was analyzed by cell counting (DAPI [4',6-diamidino-2-phenylindole] staining) and the MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assay. In addition, cells were cultured under control conditions and osteogenic conditions (media containing 10 mM ß-glycerophosphate). Quantitative expression analysis of genes encoding the osteogenic markers alkaline phosphatase (ALP), collagen type I alpha 1 chain (COL1A1), osteoprotegerin (OPG), and osteopontin (OPN) was performed after 7 and 14 days of cultivation. Calcium deposits were stained with alizarin red. RESULTS: Our studies showed that 10 ng/ml TNF did not affect the survival and metabolic activity of PDL cells. Quantitative expression analysis revealed that long-term cultures with TNF impaired osteogenic cell fate at early and late developmental stages. Furthermore, TNF significantly reduced matrix secretion in PDL cells. CONCLUSION: The present data confirm TNF as a regulatory factor of proinflammatory remodeling that influences the differentiation behavior but not the metabolism and cell proliferation of the periodontium. Therefore, TNF represents an interesting target for the regulation of orthodontic remodeling processes in the periodontium.

2.
Biomaterials ; 312: 122719, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39088912

RÉSUMÉ

Acute myeloid leukemia (AML) is a deadly form of leukemia with ineffective traditional treatment and frequent chemoresistance-associated relapse. Personalized drug screening holds promise in identifying optimal regimen, nevertheless, primary AML cells undergo spontaneous apoptosis during cultures, invalidating the drug screening results. Here, we reconstitute a 3D osteogenic niche (3DON) mimicking that in bone marrow to support primary AML cell survival and phenotype maintenance in cultures. Specifically, 3DON derived from osteogenically differentiated mesenchymal stem cells (MSC) from healthy and AML donors are co-cultured with primary AML cells. The AML cells under the AML_3DON niche showed enhanced viability, reduced apoptosis and maintained CD33+ CD34-phenotype, associating with elevated secretion of anti-apoptotic cytokines in the AML_3DON niche. Moreover, AML cells under the AML_3DON niche exhibited low sensitivity to two FDA-approved chemotherapeutic drugs, further suggesting the physiological resemblance of the AML_3DON niche. Most interestingly, AML cells co-cultured with the healthy_3DON niche are highly sensitive to the same sample drugs. This study demonstrates the differential responses of AML cells towards leukemic and healthy bone marrow niches, suggesting the impact of native cancer cell niche in drug screening, and the potential of re-engineering healthy bone marrow niche in AML patients as chemotherapeutic adjuvants overcoming chemoresistance, respectively.

3.
J Biomed Mater Res A ; 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39087511

RÉSUMÉ

Degradable phosphate glasses have shown favorable properties for tissue engineering. By changing the composition of the glasses, the degradation rate, and ion release are controllable. Zinc oxide can function as a glass network modifier and has been shown to play a positive role in bone formation. Also, phosphate glasses can easily be processed into microspheres, which can be used as microcarriers. This study aims to develop zinc phosphate glasses microspheres and explore the optimized size and composition for applications in bone tissue engineering. Zinc-titanium-calcium-sodium phosphate glasses with 0, 1, 3, 5, or 10 mol % zinc oxide were prepared and processed into microspheres. The smaller microspheres ranged in size from 50 to 106 µm, while the larger ones ranged from 106 to 150 µm. The characteristics of glasses were examined. The osteoblastic cell line MC3T3-E1 was cultured on the surface of microspheres and the cell viability was examined. To evaluate osteogenic differentiation, Alizarin Red S staining, quantitative reverse transcription polymerase chain reaction, and western blot analysis were performed after 14 days. Different sizes of zinc phosphate glass microspheres were successfully made. The glass microspheres with <10 mol % zinc oxide were able to support the adhesion and proliferation of MC3T3-E1 cell lines. The relative gene expression of BMP2 was significantly upregulated in the smaller glass microspheres containing 3 mol % zinc oxide (26-fold, p < .001) and both sizes of microspheres containing 5 mol % zinc oxide (smaller: 27-fold, p < .001; larger: 35-fold, p < .001). Additionally, cluster formation was observed in glass microspheres after 14 days, and the mineralization of MC3T3-E1 cell lines was promoted. Based on these findings, the glass microspheres containing 3-5 mol % of zinc oxide can promote osteogenic differentiation for MC3T3-E1 cells.

4.
Biomed Pharmacother ; 178: 117231, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39094542

RÉSUMÉ

AIMS: To investigate the molecular mechanism of osteoclast-derived exosomes in osteoporosis. MAIN METHODS: RANKL induced osteoclast model was screened for significantly differentially expressed lncRNAs and mRNAs by whole RNA sequencing. Exosomes were characterized using electron microscopy, western blotting and nanosight. Overexpression or knockdown of AW011738 was performed to explore its function. The degree of osteoporosis in an osteoporosis model was assessed by mirco-CT. The osteoclast model, osteoblast differentiation ability and the molecular mechanism of lncRNA AW011738/miR-24-2-5p/TREM1 axis in osteoporosis were assessed by dual luciferase reporter gene assay, Western blotting (WB), immunofluorescence and ALP staining. Bioinformatics was used to predict interactions of key osteoporosis-related genes with miRNAs, transcription factors, and potential drugs after upregulation of AW011738. To predict the protein-protein interaction (PPI) network associated with key genes, GO and KEGG analyses were performed on the key genes. The ssGSVA was used to predict changes in the immune microenvironment. KEY FINDINGS: Osteoclast-derived exosomes containing lncRNA AW011738 decreased the osteogenesis-related markers and accelerated bone loss in OVX mice. Osteoclast (si-AW011738)-derived exosomes showed a significant increase in biomarkers of osteoblast differentiation in vitro compared to the si-NC group. As analyzed by mirco-CT, tail vein injected si-AW011738 OVX mice were less osteoporotic than the control group. AW011738 inhibited osteoblast differentiation by regulating TREM1 expression through microRNA. Meanwhile, overexpression of miR-24-2-5p inhibited TREM1 expression to promote osteoblast differentiation. SIGNIFICANCE: Osteoclast-derived exosomes containing lncRNA AW011738 inhibit osteogenesis in MC3T3-E1 cells through the lncRNA AW011738/miR-24-2-5p/TREM1 axis and exacerbate osteoporosis in OVX mice.

5.
Int J Biol Macromol ; 277(Pt 2): 134338, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39089539

RÉSUMÉ

Sodium alginate (SA) has gained widespread acclaim as a carrier medium for three-dimensional (3D) bioprinting of cells and a diverse array of bioactive substances, attributed to its remarkable biocompatibility and affordability. The conventional approach for fabricating alginate-based tissue engineering constructs entails a post-treatment phase employing a calcium ion solution. However, this method proves ineffectual in addressing the predicament of low precision during the 3D printing procedure and is unable to prevent issues such as non-uniform alginate gelation and substantial distortions. In this study, we introduced borate bioactive glass (BBG) into the SA matrix, capitalizing on the calcium ions released from the degradation of BBG to incite the cross-linking reaction within SA, resulting in the formation of BBG-SA hydrogels. Building upon this fundamental concept, it unveiled that BBG-SA hydrogels greatly enhance the precision of SA in extrusion-based 3D printing and significantly reduce volumetric contraction shrinkage post-printing, while also displaying certain adhesive properties and electrical conductivity. Furthermore, in vitro cellular experiments have unequivocally established the excellent biocompatibility of BBG-SA hydrogel and its capacity to actively stimulate osteogenic differentiation. Consequently, BBG-SA hydrogel emerges as a promising platform for 3D bioprinting, laying the foundation for the development of flexible, biocompatible electronic devices.

6.
Article de Anglais | MEDLINE | ID: mdl-39044386

RÉSUMÉ

The treatment of critical-sized bone defects caused by tumor removal, skeletal injuries, or infections continues to pose a major clinical challenge. A popular potential alternative solution to autologous bone grafts is a tissue-engineered approach that utilizes the combination of mesenchymal stromal/stem cells (MSCs) with synthetic biomaterial scaffolds. This approach aims to support new bone formation by mimicking many of the biochemical and biophysical cues present within native bone. Regrettably, osteocyte cells, crucial for bone maturation and homeostasis, are rarely produced within MSC-seeded scaffolds, thereby restricting the development of fully mature cortical bone from these synthetic implants. In this work, we have constructed a multimodal scaffold by combining electrospun poly(lactic-co-glycolic acid) (PLGA) fibrous scaffolds with poly(ethylene glycol) (PEG)-based hydrogels that mimic the functional unit of cortical bone, osteon (osteon-mimetic) scaffolds. These scaffolds were decorated with a novel bone morphogenic protein-6 (BMP6) peptide (BMP6p) after our findings revealed that the BMP6p drives higher levels of Smad signaling than the full-length protein counterpart, soluble or when bound to the PEG hydrogel backbone. We show that our osteon-mimetic scaffolds, in presenting concentric layers of BMP6p-PEG hydrogel overlaid on MSC-seeded PLGA nanofibers, promoted the rapid formation of osteocyte-like cells with a phenotypic dendritic morphology, producing early osteocyte markers, including E11/gp38 (E11). Maturation of these osteocyte-like cells was further confirmed by the observation of significant dentin matrix protein 1 (DMP1) throughout our bilayered scaffolds after 3 weeks, even when cultured in a medium without dexamethasone (DEX) or any other osteogenic supplements. These results demonstrate that these osteon-mimetic scaffolds, in presenting biochemical and topographical cues reminiscent of the forming osteon, can drive the formation of osteocyte-like cells in vitro from hBMSCs without the need for any osteogenic factor media supplementation.

7.
ACS Appl Mater Interfaces ; 16(29): 37707-37721, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39001812

RÉSUMÉ

The utilization of micronano composite scaffolds has been extensively demonstrated to confer the superior advantages in bone repair compared to single nano- or micron-sized scaffolds. Nevertheless, the enhancement of bioactivities within these composite scaffolds remains challenging. In this study, we propose a novel approach to combine melt electrowriting (MEW) and solution electrospinning (SES) techniques for the fabrication of a composite scaffold incorporating hydroxyapatite (HAP), an osteogenic component, and roxithromycin (ROX), an antibacterial active component. Scanning electron microscopy (SEM) and Fourier-transform infrared spectroscopy (FTIR) confirmed the hierarchical architecture of the nanofiber-microgrid within the scaffold, as well as the successful loading of HAP and ROX. The incorporation of HAP enhanced the water absorption capacity of the composite scaffold, thus promoting cell adhesion and proliferation, as well as osteogenic differentiation. Furthermore, ROX resulted in effective antibacterial capability without any observable cytotoxicity. Finally, the scaffolds were applied to a rat calvarial defect model, and the results demonstrated that the 20% HAP group exhibited superior new bone formation without causing adverse reactions. Therefore, our findings present a promising strategy for designing and fabricating bioactive scaffolds for bone regeneration.


Sujet(s)
Antibactériens , Durapatite , Ostéogenèse , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Antibactériens/pharmacologie , Antibactériens/composition chimique , Animaux , Structures d'échafaudage tissulaires/composition chimique , Ostéogenèse/effets des médicaments et des substances chimiques , Rats , Durapatite/composition chimique , Durapatite/pharmacologie , Régénération osseuse/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Roxithromycine/composition chimique , Roxithromycine/pharmacologie , Nanofibres/composition chimique , Staphylococcus aureus/effets des médicaments et des substances chimiques , Os et tissu osseux/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris
8.
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
9.
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
10.
Curr Issues Mol Biol ; 46(7): 6346-6365, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39057021

RÉSUMÉ

Osteoporosis represents a widespread and debilitating chronic bone condition that is increasingly prevalent globally. Its hallmark features include reduced bone density and heightened fragility, which significantly elevate the risk of fractures due to the decreased presence of mature osteoblasts. The limitations of current pharmaceutical therapies, often accompanied by severe side effects, have spurred researchers to seek alternative strategies. Adipose-derived stem cells (ADSCs) hold considerable promise for tissue repair, albeit they encounter obstacles such as replicative senescence in laboratory conditions. In comparison, employing ADSCs within three-dimensional (3D) environments provides an innovative solution, replicating the natural extracellular matrix environment while offering a controlled and cost-effective in vitro platform. Moreover, the utilization of photobiomodulation (PBM) has emerged as a method to enhance ADSC differentiation and proliferation potential by instigating cellular stimulation and facilitating beneficial performance modifications. This literature review critically examines the shortcomings of current osteoporosis treatments and investigates the potential synergies between 3D cell culture and PBM in augmenting ADSC differentiation towards osteogenic lineages. The primary objective of this study is to assess the efficacy of combined 3D environments and PBM in enhancing ADSC performance for osteoporosis management. This research is notably distinguished by its thorough scrutiny of the existing literature, synthesis of recent advancements, identification of future research trajectories, and utilization of databases such as PubMed, Scopus, Web of Science, and Google Scholar for this literature review. Furthermore, the exploration of biomechanical and biophysical stimuli holds promise for refining treatment strategies. The future outlook suggests that integrating PBM with ADSCs housed within 3D environments holds considerable potential for advancing bone regeneration efforts. Importantly, this review aspires to catalyse further advancements in combined therapeutic strategies for osteoporosis regeneration.

11.
Adv Med Sci ; 69(2): 339-348, 2024 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-39004219

RÉSUMÉ

PURPOSE: The process of osteogenic differentiation hinges upon the pivotal role of mechanical signals. Previous studies found that mechanical tensile strain of 2500 microstrain (µÎµ) at a frequency of 0.5 â€‹Hz promoted osteogenesis in vitro. However, the mechanism of the mechanical strain influencing osteogenesis at the cellular and molecular levels are not yet fully understood. This study aimed to explore the mechanism of mechanical strain on osteogenic differentiation of MC3T3-E1 cells. MATERIALS AND METHODS: Proteomics analysis was conducted to explore the mechanical strain that significantly impacted the protein expression. Bioinformatics identified important mechanosensitive proteins and the expression of genes was investigated using real-time PCR. The dual-luciferase assay revealed the relationship between the miRNA and its target gene. Overexpression and downexpression of the gene, to explore its role in mechanically induced osteogenic differentiation and transcriptomics, revealed further mechanisms in this process. RESULTS: Proteomics and bioinformatics identified an important mechanosensitive lowexpression protein ATP13A3, and the expression of Atp13a3 gene was also reduced. The dual-luciferase assay revealed that microRNA-3070-3p (miR-3070-3p) targeted the Atp13a3 gene. Furthermore, the downexpression of Atp13a3 promoted the expression levels of osteogenic differentiation-related genes and proteins, and this process was probably mediated by the tumor necrosis factor (TNF) signaling pathway. CONCLUSION: Atp13a3 responded to mechanical tensile strain to regulate osteogenic differentiation, and the TNF signaling pathway regulated by Atp13a3 was probably involved in this process. These novel insights suggested that Atp13a3 was probably a potential osteogenesis and bone formation regulator.

12.
Bone ; : 117197, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38986825

RÉSUMÉ

Bone marrow mesenchymal stem cells (BMSCs) are integral to bone remodeling and homeostasis, as they are capable of differentiating into osteogenic and adipogenic lineages. This differentiation is substantially influenced by mechanosensitivity, particularly to tensile strain, which is a prevalent mechanical stimulus known to enhance osteogenic differentiation. This review specifically examines the effects of various cyclic tensile stress (CTS) conditions on BMSC osteogenesis. It delves into the effects of different loading devices, magnitudes, frequencies, elongation levels, dimensionalities, and coculture conditions, providing a comparative analysis that aids identification of the most conducive parameters for the osteogenic differentiation of BMSCs. Subsequently, this review delineates the signaling pathways activated by CTS, such as Wnt/ß-catenin, BMP, Notch, MAPK, PI3K/Akt, and Hedgehog, which are instrumental in mediating the osteogenic differentiation of BMSCs. Through a detailed examination of these pathways, this study elucidates the intricate mechanisms whereby tensile strain promotes osteogenic differentiation, offering valuable guidance for optimizing therapeutic strategies aimed at enhancing bone regeneration.

13.
ACS Appl Mater Interfaces ; 16(28): 36017-36029, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38975983

RÉSUMÉ

Oral infectious diseases have a significant impact on the health of oral and maxillofacial regions, as well as the overall well-being of individuals. Carvacrol and thymol, two isomers known for their effective antibacterial and anti-inflammatory properties, have gained considerable attention in the treatment of oral infectious diseases. However, their application as topical drugs for oral use is limited due to their poor physical and chemical stability. UiO-66, a metal-organic framework based on zirconium ion (Zr4+), exhibits high drug loading capability. Carvacrol and thymol were efficiently loaded onto UiO-66 with loading rates of 79.60 ± 0.71% and 79.65 ± 0.76%, respectively. The release rates of carvacrol and thymol were 77.82 ± 0.87% and 76.51 ± 0.58%, respectively, after a period of 72 h. Moreover, Car@UiO-66 and Thy@UiO-66 demonstrated excellent antibacterial properties against Candida albicans, Escherichia coli, and Staphylococcus aureus with minimum bactericidal concentrations (MBC) of 0.313 mg/mL, 0.313 mg/mL, and 1.25 mg/mL, respectively. Furthermore, based on the results of the CCK8 cytotoxicity assay, even at concentrations as high as 1.25 mg/mL, Car@UiO-66 and Thy@UiO-66 exhibited excellent biocompatibility with a relative cell survival rate above 50%. These findings suggest that Car@UiO-66 and Thy@UiO-66 possess favorable biocompatibility properties without significant toxicity towards periodontal membrane cells. Additionally, in vivo studies confirmed the efficacy of Car@UiO-66and Thy@UiO-66 in reducing inflammation, promoting bone formation through inhibition of TNF-a and IL6 expression, enhancement of IL10 expression, and acceleration of bone defect healing. Therefore, the unique combination of antibacterial, anti-inflammatory, and osteogenic properties make Car@UiO-66 and Thy@Ui O-66 promising candidates for the treatment of oral infectious diseases and repairing bone defects.


Sujet(s)
Antibactériens , Anti-inflammatoires , Candida albicans , Cymènes , Escherichia coli , Réseaux organométalliques , Staphylococcus aureus , Thymol , Thymol/composition chimique , Thymol/pharmacologie , Cymènes/composition chimique , Cymènes/pharmacologie , Antibactériens/pharmacologie , Antibactériens/composition chimique , Animaux , Anti-inflammatoires/composition chimique , Anti-inflammatoires/pharmacologie , Staphylococcus aureus/effets des médicaments et des substances chimiques , Escherichia coli/effets des médicaments et des substances chimiques , Candida albicans/effets des médicaments et des substances chimiques , Réseaux organométalliques/composition chimique , Réseaux organométalliques/pharmacologie , Souris , Tests de sensibilité microbienne , Rats , Ostéogenèse/effets des médicaments et des substances chimiques , Humains
14.
Front Genet ; 15: 1429844, 2024.
Article de Anglais | MEDLINE | ID: mdl-39015772

RÉSUMÉ

Mesenchymal stem cells (MSCs) have promising potential for bone tissue engineering in bone healing and regeneration. They are regarded as such due to their capacity for self-renewal, multiple differentiation, and their ability to modulate the immune response. However, changes in the molecular pathways and transcription factors of MSCs in osteogenesis can lead to bone defects and metabolic bone diseases. DNA methylation is an epigenetic process that plays an important role in the osteogenic differentiation of MSCs by regulating gene expression. An increasing number of studies have demonstrated the significance of DNA methyltransferases (DNMTs), Ten-eleven translocation family proteins (TETs), and MSCs signaling pathways about osteogenic differentiation in MSCs. This review focuses on the progress of research in these areas.

15.
Cureus ; 16(6): e62351, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39006559

RÉSUMÉ

BACKGROUND: Stem cells of mesenchymal origin have good proliferative capacity when compared to other stem cell types. Dental pulp stem cells (DPSCs) are a variety of mesenchymal cells obtained from the pulpal tissue of teeth and are abundantly available and easy to obtain. DPSCs facilitate and improve the formation of new bone using different bone graft scaffolds. This present study aims to evaluate and compare the osteogenic potential of DPSCs on alloplastic and xenogeneic bone grafts. MATERIALS AND METHODS: Hydroxyapatite and beta-tricalcium bone graft and bovine bone graft were used in a triplicate manner in the laboratory. DPSCs were obtained from the pulpal tissue of extracted third molars in the laboratory. The cytotoxicity, osteogenic potential, and difference in the rate of proliferation of mesenchymal cells on the biomaterials were assessed. RESULTS: Darker purple staining was seen in the case of hydroxyapatite/beta-tricalcium bone graft on MTT colorimetric assay stating that there was an increase in cell viability in hydroxyapatite/beta-tricalcium bone graft as compared to the bovine bone graft. Hydroxyapatite/beta-tricalcium bone graft showed more osteogenic potential as compared to the bovine bone graft as a higher degree of red staining was seen in Alizarin staining. CONCLUSION: Higher cell viability and higher osteogenic proliferation and differentiation were seen on the hydroxyapatite/beta-tricalcium bone graft compared to the bovine bone scaffold.

16.
Mol Biol Rep ; 51(1): 838, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39042226

RÉSUMÉ

BACKGROUND: Bioglass materials have gained significant attention in the field of tissue engineering due to their osteoinductive and biocompatible properties that promote bone cell differentiation. In this study, a novel composite scaffold was developed using a sol-gel technique to combine bioglass (BG) 58 S with a poly L-lactic acid (PLLA). METHODS AND RESULTS: The physiochemical properties, morphology, and osteoinductive potential of the scaffolds were investigated by X-ray diffraction analysis, scanning electron microscopy, and Fourier-transform infrared spectroscopy. The results showed that the SiO2-CaO-P2O5 system was successfully synthesized by the sol-gel method. The PLLA scaffolds containing BG was found to be osteoinductive and promoted mineralization, as demonstrated by calcium deposition assay, upregulation of alkaline phosphatase enzyme activity, and Alizarin red staining data. CONCLUSIONS: These in vitro studies suggest that composite scaffolds incorporating hBMSCs are a promising substitute material to be implemented in bone tissue engineering. The PLLA/BG scaffolds promote osteogenesis and support the differentiation of bone cells, such as osteoblasts, due to their osteoinductive properties.


Sujet(s)
Matériaux biocompatibles , Différenciation cellulaire , Céramiques , Ostéogenèse , Polyesters , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Polyesters/composition chimique , Structures d'échafaudage tissulaires/composition chimique , Céramiques/composition chimique , Céramiques/pharmacologie , Ingénierie tissulaire/méthodes , Matériaux biocompatibles/pharmacologie , Matériaux biocompatibles/composition chimique , Ostéogenèse/effets des médicaments et des substances chimiques , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Régénération osseuse/effets des médicaments et des substances chimiques , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Spectroscopie infrarouge à transformée de Fourier , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Diffraction des rayons X , Os et tissu osseux/effets des médicaments et des substances chimiques , Os et tissu osseux/métabolisme , Phosphatase alcaline/métabolisme , Microscopie électronique à balayage
17.
J Orthop Surg Res ; 19(1): 386, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951811

RÉSUMÉ

BACKGROUND: Bone defects, resulting from substantial bone loss that exceeds the natural self-healing capacity, pose significant challenges to current therapeutic approaches due to various limitations. In the quest for alternative therapeutic strategies, bone tissue engineering has emerged as a promising avenue. Notably, excretory proteins from Toxoplasma gondii (TgEP), recognized for their immunogenicity and broad spectrum of biological activities secreted or excreted during the parasite's lifecycle, have been identified as potential facilitators of osteogenic differentiation in human bone marrow mesenchymal stem cells (hBMSCs). Building on our previous findings that TgEP can enhance osteogenic differentiation, this study investigated the molecular mechanisms underlying this effect and assessed its therapeutic potential in vivo. METHODS: We determined the optimum concentration of TgEP through cell cytotoxicity and cell proliferation assays. Subsequently, hBMSCs were treated with the appropriate concentration of TgEP. We assessed osteogenic protein markers, including alkaline phosphatase (ALP), Runx2, and Osx, as well as components of the BMP/Smad signaling pathway using quantitative real-time PCR (qRT-PCR), siRNA interference of hBMSCs, Western blot analysis, and other methods. Furthermore, we created a bone defect model in Sprague-Dawley (SD) male rats and filled the defect areas with the GelMa hydrogel, with or without TgEP. Microcomputed tomography (micro-CT) was employed to analyze the bone parameters of defect sites. H&E, Masson and immunohistochemical staining were used to assess the repair conditions of the defect area. RESULTS: Our results indicate that TgEP promotes the expression of key osteogenic markers, including ALP, Runx2, and Osx, as well as the activation of Smad1, BMP2, and phosphorylated Smad1/5-crucial elements of the BMP/Smad signaling pathway. Furthermore, in vivo experiments using a bone defect model in rats demonstrated that TgEP markedly promoted bone defect repair. CONCLUSION: Our results provide compelling evidence that TgEP facilitates hBMSC osteogenic differentiation through the BMP/Smad signaling pathway, highlighting its potential as a therapeutic approach for bone tissue engineering for bone defect healing.


Sujet(s)
Différenciation cellulaire , Cellules souches mésenchymateuses , Ostéogenèse , Rat Sprague-Dawley , Transduction du signal , Toxoplasma , Cellules souches mésenchymateuses/métabolisme , Ostéogenèse/physiologie , Humains , Animaux , Transduction du signal/physiologie , Différenciation cellulaire/physiologie , Mâle , Toxoplasma/physiologie , Rats , Protéines Smad/métabolisme , Protéines de protozoaire/métabolisme , Protéines morphogénétiques osseuses/métabolisme , Cellules cultivées
18.
Int Endod J ; 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38973098

RÉSUMÉ

AIM: The regenerative capacity of dental pulp relies on the odonto/osteogenic differentiation of dental pulp cells (DPCs), but dynamic microenvironmental changes hinder the process. Bone morphogenetic protein 9 (BMP9) promotes differentiation of DPCs towards an odonto/osteogenic lineage, forming dentinal-like tissue. However, the molecular mechanism underlying its action remains unclear. This study investigates the role of DLX6 antisense RNA 1 (DLX6-AS1) in odonto/osteogenic differentiation induced by BMP9. METHODOLOGY: Custom RT2 profiler PCR array, quantitative Real-Time PCR (qRT-PCR) and western blots were used to investigate the expression pattern of DLX6-AS1 and its potential signal axis. Osteogenic ability was evaluated using alkaline phosphatase and alizarin red S staining. Interactions between lncRNA and miRNA, as well as miRNA and mRNA, were predicted through bioinformatic assays, which were subsequently validated via RNA immunoprecipitation and dual luciferase reporter assays. Student's t-test or one-way ANOVA with post hoc Tukey HSD tests were employed for data analysis, with a p-value of less than .05 considered statistically significant. RESULTS: DLX6-AS1 was upregulated upon BMP9 overexpression in DPCs, thereby promoting odonto/osteogenic differentiation. Additionally, miR-128-3p participated in BMP9-induced odonto/osteogenic differentiation by interacting with the downstream signal MAPK14. Modifying the expression of miR-128-3p and transfecting pcMAPK14/siMAPK14 had a rescue impact on odonto/osteogenic differentiation downstream of DLX6-AS1. Lastly, miR-128-3p directly interacted with both MAPK14 and DLX6-AS1. CONCLUSIONS: DLX6-AS1 could regulate the odonto/osteogenic differentiation of DPCs under the control of BMP9 through the miR-128-3p/MAPK14 axis.

19.
Biomed Mater ; 19(5)2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38955344

RÉSUMÉ

Artificial bone substitutes for bone repair and reconstruction still face enormous challenges. Previous studies have shown that calcium magnesium phosphate cements (CMPCs) possess an excellent bioactive surface, but its clinical application is restricted due to short setting time. This study aimed to develop new CMPC/carboxymethyl chitosan (CMCS) comg of mixed powders of active MgO, calcined MgO and calcium dihydrogen phosphate monohydrate. With this novel strategy, it can adjust the setting time and improve the compressive strength. The results confirmed that CMPC/CMCS composite bone cements were successfully developed with a controllable setting time (18-70 min) and high compressive strength (87 MPa). In addition, the composite bone cements could gradually degrade in PBS with weight loss up to 32% at 28 d. They also promoted the proliferation of pre-osteoblasts, and induced osteogenic differentiation. The findings indicate that CMPC/CMCS composite bone cements hold great promise as a new type of bone repair material in further and in-depth studies.


Sujet(s)
Matériaux biocompatibles , Ciments osseux , Phosphates de calcium , Différenciation cellulaire , Prolifération cellulaire , Chitosane , Résistance à la compression , Composés du magnésium , Test de matériaux , Ostéoblastes , Ostéogenèse , Chitosane/composition chimique , Chitosane/analogues et dérivés , Ciments osseux/composition chimique , Ciments osseux/pharmacologie , Ostéogenèse/effets des médicaments et des substances chimiques , Phosphates de calcium/composition chimique , Phosphates de calcium/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/cytologie , Composés du magnésium/composition chimique , Composés du magnésium/pharmacologie , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Substituts osseux/composition chimique , Substituts osseux/pharmacologie , Phosphates
20.
Acta Biomater ; 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38997078

RÉSUMÉ

Biodegradable Zn alloys have significant application potential for hard-tissue implantation devices owing to their suitable degradation behavior and favorable biocompatibility. Nonetheless, pure Zn and its alloys in the as-cast state are mechanically instable and low in strength, which restricts their clinical applicability. Here, we report the exceptional mechanical, corrosion, and biocompatibility properties of hot-extruded Zn-5RE (wt.%, RE = rare earth of Y; or Ho; or Er) alloys intended for use in biodegradable bone substitutes. The microstructural characteristics, mechanical behavior, corrosion resistance, cytocompatibility, osteogenic differentiation, and capacity of osteogenesis in vivo of the Zn-5RE alloys are comparatively investigated. The Zn-5Y alloy demonstrates the best tensile properties, encompassing a 138 MPa tensile yield strength, a 302 MPa ultimate tensile strength, and 63% elongation, while the Zn-5Ho alloy shows the highest compression yield strength of 260 MPa and Vickers hardness of 104 HV. The Zn-5Er alloy shows a 126 MPa tensile yield strength, a 279 MPa ultimate tensile strength, 52% elongation, a 196 MPa compression yield strength, and a 101 HV Vickers microhardness. Further, the Zn-5Er alloy has a 130 µm per year corrosion rate in electrochemical tests and a 26 µm per year degradation rate in immersion tests, which is the lowest among the tested alloys. It also has the best in vitro osteogenic differentiation ability and capacity for osteogenesis and osteointegration in vivo after implantation in rat femurs among the Zn-5RE alloys, indicating promising potential in load-bearing biodegradable internal bone-fixation applications. STATEMENT OF SIGNIFICANCE: This work reports the exceptional mechanical, corrosion, and biocompatibility properties of hot-extruded (HE) Zn-5 wt.%-rare earth (Zn-5RE) alloys using single yttrium (Y), holmium (Ho), and erbium (Er) alloying for biodegradable bone-implant applications. Our findings demonstrate that the HE Zn-5Er alloy showed σuts of 279 MPa, tensile yield strength of 126 MPa, elongation of 51.6%, compression yield strength of 196 MPa, and microhardness of 101.2 HV. Further, HE Zn-5Er showed the lowest electrochemical corrosion rate of 130 µm/y and lowest degradation rate of 26 µm/y, and the highest in vitro osteogenic differentiation ability, in vivo osteogenesis, and osteointegration ability after implantation in rat femurs among the Zn-5RE alloys, indicating promising potential in load-bearing biodegradable internal bone-fixation applications.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE