Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biol Direct ; 19(1): 28, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38650011

ABSTRACT

BACKGROUND: Osteosarcoma is a diverse and aggressive bone tumor. Driver genes regulating osteosarcoma initiation and progression remains incompletely defined. Zinc finger protein 692 (ZNF692), a kind of Krüppel C2H2 zinc finger transcription factor, exhibited abnormal expression in different types of malignancies and showed a correlation with the clinical prognosis of patients as well as the aggressive characteristics of cancer cells. Nevertheless, its specific role in osteosarcoma is still not well understood. METHODS: We investigated the dysregulation and clinical significance of ZNF692 in osteosarcoma through bioinformatic method and experimental validation. A range of in vitro assays, including CCK-8, colony formation, EdU incorporation, wound healing, and transwell invasion tests, were conducted to assess the impact of ZNF692 on cell proliferation, migration, and invasion in osteosarcoma. A xenograft mouse model was established to evaluate the effect of ZNF692 on tumor growth in vivo. Western blot assay was used to measure the protein levels of MEK1/2, P-MEK1/2, ERK1/2, and P-ERK1/2 in cells that had been genetically modified to either reduce or increase the expression of ZNF692. The relationship between ZNF692 and tyrosine kinase non-receptor 2 (TNK2) were validated by qRT-PCR, chromatin immunoprecipitation and luciferase reporter assays. RESULTS: Expression of ZNF692 was increased in both human osteosarcoma tissues and cell lines. Furthermore, the expression of ZNF692 served as an independent predictive biomarker in osteosarcoma. The results of the survival analysis indicated that increased expression of ZNF692 was associated with worse outcome. Downregulation of ZNF692 inhibits the proliferation, migration, and invasion of osteosarcoma cells, whereas upregulation of ZNF692 has the opposite impact. Western blot assay indicates that reducing ZNF692 decreases phosphorylation of MEK1/2 and ERK1/2, whereas increasing ZNF692 expression enhances their phosphorylation. U0126, a potent inhibitor specifically targeting the MEK/ERK signaling pathway, partially counteracts the impact of ZNF692 overexpression on the proliferation, migration, and invasion of osteosarcoma cells. In addition, ZNF692 specifically interacts with the promoter region of TNK2 and stimulates the transcription of TNK2 in osteosarcoma cells. Forcing the expression of TNK2 weakens the inhibitory impact of ZNF692 knockdown on P-MEK1/2 and P-ERK1/2. Similarly, partly inhibiting TNK2 counteracts the enhancing impact of ZNF692 overexpression on the phosphorylation of MEK1/2 and ERK1/2. Functional tests demonstrate that the suppressive effects of ZNF692 knockdown on cell proliferation, migration, and invasion are greatly reduced when TNK2 is overexpressed. In contrast, the reduction of TNK2 hinders the ability of ZNF692 overexpression to enhance cell proliferation, migration, and invasion. CONCLUSION: ZNF692 promotes the proliferation, migration, and invasion of osteosarcoma cells via the TNK2-dependent stimulation of the MEK/ERK signaling pathway. The ZNF692-TNK2 axis might potentially function as a possible predictive biomarker and a promising target for novel therapeutics in osteosarcoma.


Subject(s)
Cell Movement , Cell Proliferation , MAP Kinase Signaling System , Osteosarcoma , Animals , Female , Humans , Mice , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Mice, Nude , Neoplasm Invasiveness , Osteosarcoma/genetics , Osteosarcoma/metabolism
2.
Int J Biol Macromol ; 266(Pt 1): 130998, 2024 May.
Article in English | MEDLINE | ID: mdl-38521332

ABSTRACT

Although calcium­magnesium phosphate cements (CMPCs) have been widely applied to treating critical-size bone defects, their repair efficiency is unsatisfactory owing to their weak surface bioactivity and uncontrolled ion release. In this study, we lyophilized alginate sodium (AS) as a coating onto HAp/K-struvite (H@KSv) to develop AS/HAp/K-struvite (AH@KSv), which promotes bone regeneration. The compressive strength and hydrophilicity of AH@KSv significantly improved, leading to enhanced cell adhesion in vitro. Importantly, the SA coating enables continuous ions release of Mg2+ and Ca2+, finally leading to enhanced osteogenesis in vitro/vivo and different patterns of new bone ingrowth in vivo. Furthermore, these composites increased the expression levels of biomarkers of the TRPM7/PI3K/Akt signaling pathway via an equilibrium effect of Mg2+ to Ca2+. In conclusion, our study provides novel insights into the mechanisms of Mg-based biomaterials for bone regeneration.


Subject(s)
Alginates , Bone Cements , Bone Regeneration , Phosphates , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TRPM Cation Channels , Bone Regeneration/drug effects , TRPM Cation Channels/metabolism , Alginates/chemistry , Alginates/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Phosphates/chemistry , Phosphates/pharmacology , Bone Cements/chemistry , Bone Cements/pharmacology , Osteogenesis/drug effects , Magnesium Compounds/chemistry , Magnesium Compounds/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Cell Adhesion/drug effects , Surface Properties , Mice , Rats , Compressive Strength
3.
Biomedicines ; 12(1)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38275399

ABSTRACT

Maintaining proper mechanical strength and tissue volume is important for bone growth at the site of a bone defect. In this study, potassium magnesium phosphate hexahydrate (KMgPO4·6H2O, MPC) was applied to gelma-methacrylate hydrogel (GelMA) to prepare GelMA/MPC composites (GMPCs). Among these, 5 GMPC showed the best performance in vivo and in vitro. These combinations significantly enhanced the mechanical strength of GelMA and regulated the degradation and absorption rate of MPC. Considerably better mechanical properties were noted in 5 GMPC compared with other concentrations. Better bioactivity and osteogenic ability were also found in 5 GMPC. Magnesium ions (Mg2+) are bioactive and proven to promote bone tissue regeneration, in which the enhancement efficiency is closely related to Mg2+ concentrations. These findings indicated that GMPCs that can release Mg2+ are effective in the treatment of bone defects and hold promise for future in vivo applications.

4.
Mol Med ; 29(1): 133, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37789274

ABSTRACT

BACKGROUND: Rab-interacting lysosomal protein (RILP) contains an alpha-helical coil with an unexplored biological function in osteosarcoma. This study investigated the expression of RILP in osteosarcoma cells and tissues to determine the effect of RILP on the biological behaviors of osteosarcoma cells and the underlying mechanism. METHODS: Tumor Immune Estimation Resource (TIMER) database, The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database were used for bioinformatic analysis. Co-immunoprecipitation experiment was used to determine whether the two proteins were interacting. In functional tests, cell counting kit-8 (CCK-8) assay, colony formation assay, wound healing assay, transwell invasion assay, Immunofluorescence (IF) assay and immunohistochemical (IHC) assay were performed. RESULTS: Overexpression of RILP significantly inhibited proliferation and impaired metastasis ability of osteosarcoma cells, while silencing of RILP showed the opposite trend. RNA-seq data analysis was applied in 143B cells and pathway enrichment analysis revealed that differentially expressed genes were mainly enriched in the PI3K/AKT pathway. We further verified that overexpression of RILP restrained the PI3K/AKT/mTOR signaling pathway and induced autophagy in osteosarcoma cells, while the opposite trend was observed when PI3K pathway activator 740Y-P was used. 3-Methyladenine (3-MA), a selective autophagy inhibitor, partially attenuated the inhibitory effect of RILP on the migration and invasion ability of osteosarcoma cells, suggesting the involvement of autophagy in epithelial-mesenchymal transition regulation in osteosarcoma cells. Growth factor receptor binding protein-10 (Grb10), an adaptor protein, was confirmed as a potential target of RILP to restrain the PI3K/AKT signaling pathway. We subcutaneously injected stably overexpressing 143B osteosarcoma cells into nude mice and observed that overexpression of RILP inhibited tumor growth by inhibiting the PI3K/AKT/mTOR pathway. CONCLUSION: Our study revealed that the expression of RILP was associated with favorable prognosis of osteosarcoma and RILP inhibits proliferation, migration, and invasion and promotes autophagy in osteosarcoma cells via Grb10-mediated inhibition of the PI3K/AKT/mTOR signaling pathway. In the future, targeting RILP may be a potential strategy for osteosarcoma treatment.


Subject(s)
Bone Neoplasms , Osteosarcoma , Animals , Mice , Apoptosis , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , GRB10 Adaptor Protein/metabolism , GRB10 Adaptor Protein/pharmacology , Mice, Nude , Osteosarcoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Humans
5.
Biomedicines ; 12(1)2023 Dec 28.
Article in English | MEDLINE | ID: mdl-38255182

ABSTRACT

Bone defect repair poses significant challenges in orthopedics, thereby increasing the demand for bone substitutes. Magnesium phosphate cements (MPCs) are widely used for bone defect repair because of their excellent mechanical properties and biodegradability. However, high crystallinity and uncontrolled magnesium ion (Mg2+) release limit the surface bioactivity of MPCs in bone regeneration. Here, we fabricate chondroitin sulfate (CS) as a surface coating via the lyophilization method, namely CMPC. We find that the CS coating is uniformly distributed and improves the mechanical properties of MPC through anionic electrostatic adsorption, while mediating degradation-related controlled ion release of Mg2+. Using a combination of in vitro and in vivo analyses, we show that the CS coating maintained cytocompatibility while increasing the cell adhesion area of MC3T3-E1s. Furthermore, we display accelerated osteogenesis and angiogenesis of CMPC, which are related to appropriate ion concentration of Mg2+. Our findings reveal that the preparation of a lyophilized CS coating is an effective method to promote surface bioactivity and mediate Mg2+ concentration dependent osteogenesis and angiogenesis, which have great potential in bone regeneration.

6.
Front Cell Dev Biol ; 9: 699212, 2021.
Article in English | MEDLINE | ID: mdl-34368151

ABSTRACT

Osteosarcoma is the most common malignant bone tumor, and although there has been significant progress in its management, metastases often herald incurable disease. Here we defined genes differentially expressed between primary and metastatic osteosarcoma as metastasis-related genes (MRGs) and used them to construct a novel six-MRG prognostic signature for overall survival of patients with osteosarcoma. Validation in internal and external datasets confirmed satisfactory accuracy and generalizability of the prognostic model, and a nomogram based on the signature and clinical variables was constructed to aid clinical decision-making. Of the six MRGs, FHIT is a well-documented tumor suppressor gene that is poorly defined in osteosarcoma. Consistent with tumor suppressor function, FHIT was downregulated in osteosarcoma cells and human osteosarcoma samples. FHIT overexpression inhibited osteosarcoma proliferation, migration, and invasion both in vitro and in vivo. Mechanistically, FHIT overexpression upregulate the epithelial marker E-cadherin while repressing the mesenchymal markers N-cadherin and vimentin. Our six-MRG signature represents a novel and clinically useful prognostic biomarker for patients with osteosarcoma, and FHIT might represent a therapeutic target by reversing epithelial to mesenchymal transition.

7.
Biomed Mater ; 16(3)2021 04 16.
Article in English | MEDLINE | ID: mdl-33827063

ABSTRACT

Magnesium phosphate cement (MPC) has been evaluated as an inorganic bone filler due to its favorable biocompatibility, biodegradability, rapid setting, high initial strength, and osteogenic potential. However, the setting time of MPC is so rapid that it makes it difficult to use in practice, and the clinical properties of MPC could be further be improved by adding bioactive materials. Here we developed novel bioactive chondroitin sulfate (CS)-MPC composites (CS-MPCs) by incorporating different amounts of CS into MPC. The compositions, microstructures, and physiochemical properties of CS-MPCs and their inducedin vitrocellular responses andin vivobone regeneration properties were evaluated. CS-MPCs had a longer setting time, lower hydration temperature, higher compressive strength, and more neural pH than MPC. CS-MPCs demonstrated similar degradation ratios relative to MPC in Tris-HCl solution. CS-MPCs promoted pre-osteoblast cell proliferation, attachment, and differentiationin vitroand enhanced bone formation surrounding implantsin vivo. In conclusion, through CS modification, our novel CS-MPCs have improved physiochemical properties that enhance compatibilityin vitroand bone regenerationin vivo, making them attractive materials for bone regeneration.


Subject(s)
Bone Cements/chemistry , Bone Regeneration , Chondroitin Sulfates/chemistry , 3T3 Cells , Animals , Bone and Bones , Cell Adhesion , Cell Differentiation , Cell Proliferation , Compressive Strength , Dental Materials/chemistry , Hydrogen-Ion Concentration , Magnesium Compounds/chemistry , Materials Testing , Mesenchymal Stem Cells , Mice , Osteoblasts/cytology , Osteogenesis , Phosphates/chemistry , Porosity , Powders , Rats , Rats, Sprague-Dawley , X-Ray Microtomography
8.
Regen Biomater ; 8(1): rbaa048, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33732494

ABSTRACT

Incorporating bioactive substances into synthetic bioceramic scaffolds is challenging. In this work, oxygen-carboxymethyl chitosan (O-CMC), a natural biopolymer that is nontoxic, biodegradable and biocompatible, was introduced into magnesium potassium phosphate cement (K-struvite) to enhance its mechanical properties and cytocompatibility. This study aimed to develop O-CMC/magnesium potassium phosphate composite bone cement (OMPC), thereby combining the optimum bioactivity of O-CMC with the extraordinary self-setting properties and mechanical intensity of the K-struvite. Our results indicated that O-CMC incorporation increased the compressive strength and setting time of K-struvite and decreased its porosity and pH value. Furthermore, OMPC scaffolds remarkably improved the proliferation, adhesion and osteogenesis related differentiation of MC3T3-E1 cells. Therefore, O-CMC introduced suitable physicochemical properties to K-struvite and enhanced its cytocompatibility for use in bone regeneration.

9.
Int J Biol Macromol ; 160: 101-111, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32450325

ABSTRACT

Magnesium phosphate cement (MPC) can be injected to form an in situ scaffold to repair bone defects. Here we synthesized novel injectable bioactive cements (CMPCs) by incorporating different ratios of carboxymethyl chitosan (CMC, 0-10%) into MPC. The physiochemical properties, compositions, and microstructures of CMPCs were evaluated. The in vitro cellular responses of pre-osteoblast MC3T3-E1 cells to CMPCs including adhesion, proliferation, and differentiation were quantified and the underlying cellular mechanisms investigated. CMPCs had longer setting times and lower setting temperatures. CMPC injectability was enhanced by the addition of CMC. The CMPC containing 5% CMC had the highest compressive strength and washout resistance. CMPCs had a more neutral pH compared to MPC at four weeks. Furthermore, CMPC samples showed similar degradability and Mg2+ release to MPC in Tris-HCl buffer. Osteoblasts (MC3T3-E1) showed significantly greater adherence, proliferation, and differentiation on CMPC specimens than on MPC. Finally, CMPCs effectively increased the adsorption of fibronectin and activated integrin signaling as indicated by enhanced FAK and ERK phosphorylation. Our novel CMPC composites have improved physicochemical properties and cellular responses and represent a promising material for bone regeneration.


Subject(s)
Bone Cements/chemistry , Bone Cements/pharmacology , Bone Regeneration/drug effects , Chitosan/analogs & derivatives , Magnesium Compounds/chemistry , Magnesium Compounds/pharmacology , Phosphates/chemistry , Phosphates/pharmacology , 3T3 Cells , Animals , Biocompatible Materials/chemistry , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Chitosan/chemistry , Chitosan/pharmacology , Compressive Strength/drug effects , Fibronectins/metabolism , Focal Adhesion Kinase 1/metabolism , Hydrogen-Ion Concentration , MAP Kinase Signaling System/drug effects , Materials Testing/methods , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...