Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 12.659
Filtrar
1.
Biomaterials ; 312: 122724, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39106818

RESUMEN

The residual bone tumor and defects which is caused by surgical therapy of bone tumor is a major and important problem in clinicals. And the sequential treatment for irradiating residual tumor and repairing bone defects has wildly prospects. In this study, we developed a general modification strategy by gallic acid (GA)-assisted coordination chemistry to prepare black calcium-based materials, which combines the sequential photothermal therapy of bone tumor and bone defects. The GA modification endows the materials remarkable photothermal properties. Under the near-infrared (NIR) irradiation with different power densities, the black GA-modified bone matrix (GBM) did not merely display an excellent performance in eliminating bone tumor with high temperature, but showed a facile effect of the mild-heat stimulation to accelerate bone regeneration. GBM can efficiently regulate the microenvironments of bone regeneration in a spatial-temporal manner, including inflammation/immune response, vascularization and osteogenic differentiation. Meanwhile, the integrin/PI3K/Akt signaling pathway of bone marrow mesenchymal stem cells (BMSCs) was revealed to be involved in the effect of osteogenesis induced by the mild-heat stimulation. The outcome of this study not only provides a serial of new multifunctional biomaterials, but also demonstrates a general strategy for designing novel blacked calcium-based biomaterials with great potential for clinical use.


Asunto(s)
Neoplasias Óseas , Regeneración Ósea , Calcio , Ácido Gálico , Células Madre Mesenquimatosas , Ácido Gálico/química , Regeneración Ósea/efectos de los fármacos , Animales , Calcio/metabolismo , Neoplasias Óseas/terapia , Neoplasias Óseas/tratamiento farmacológico , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Terapia Fototérmica/métodos , Osteogénesis/efectos de los fármacos , Ratones , Humanos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Línea Celular Tumoral
2.
Biomaterials ; 313: 122756, 2025 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39182327

RESUMEN

Currently, the treatment of bone defects in arthroplasty is a challenge in clinical practice. Nonetheless, commercially available orthopaedic scaffolds have shown limited therapeutic effects for large bone defects, especially for massiveand irregular defects. Additively manufactured porous tantalum, in particular, has emerged as a promising material for such scaffolds and is widely used in orthopaedics for its exceptional biocompatibility, osteoinduction, and mechanical properties. Porous tantalum has also exhibited unique advantages in personalised rapid manufacturing, which allows for the creation of customised scaffolds with complex geometric shapes for clinical applications at a low cost and high efficiency. However, studies on the effect of the pore structure of additively manufactured porous tantalum on bone regeneration have been rare. In this study, our group designed and fabricated a batch of precision porous tantalum scaffolds via laser powder bed fusion (LPBF) with pore sizes of 250 µm (Ta 250), 450 µm (Ta 450), 650 µm (Ta 650), and 850 µm (Ta 850). We then performed a series of in vitro experiments and observed that all four groups showed good biocompatibility. In particular, Ta 450 demonstrated the best osteogenic performance. Afterwards, our team used a rat bone defect model to determine the in vivo osteogenic effects. Based on micro-computed tomography and histology, we identified that Ta 450 exhibited the best bone ingrowth performance. Subsequently, sheep femur and hip defect models were used to further confirm the osteogenic effects of Ta 450 scaffolds. Finally, we verified the aforementioned in vitro and in vivo results via clinical application (seven patients waiting for revision total hip arthroplasty) of the Ta 450 scaffold. The clinical results confirmed that Ta 450 had satisfactory clinical outcomes up to the 12-month follow-up. In summary, our findings indicate that 450 µm is the suitable pore size for porous tantalum scaffolds. This study may provide a new therapeutic strategy for the treatment of massive, irreparable, and protracted bone defects in arthroplasty.


Asunto(s)
Regeneración Ósea , Tantalio , Andamios del Tejido , Tantalio/química , Regeneración Ósea/efectos de los fármacos , Porosidad , Animales , Andamios del Tejido/química , Ratas , Ratas Sprague-Dawley , Osteogénesis/efectos de los fármacos , Humanos , Masculino , Prueba de Estudio Conceptual , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Femenino
3.
Biomaterials ; 313: 122772, 2025 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39190942

RESUMEN

Implant-associated infection (IAI) has become an intractable challenge in clinic. The healing of IAI is a complex physiological process involving a series of spatiotemporal connected events. However, existing titanium-based implants in clinic suffer from poor antibacterial effect and single function. Herein, a versatile surface platform based on the presentation of sequential function is developed. Fabrication of titania nanotubes and poly-γ-glutamic acid (γ-PGA) achieves the efficient incorporation of silver ions (Ag+) and the pH-sensitive release in response to acidic bone infection microenvironment. The optimized PGA/Ag platform exhibits satisfactory biocompatibility and converts macrophages from pro-inflammatory M1 to pro-healing M2 phenotype during the subsequent healing stage, which creates a beneficial osteoimmune microenvironment and promotes angio/osteogenesis. Furthermore, the PGA/Ag platform mediates osteoblast/osteoclast coupling through inhibiting CCL3/CCR1 signaling. These biological effects synergistically improve osseointegration under bacterial infection in vivo, matching the healing process of IAI. Overall, the novel integrated PGA/Ag surface platform proposed in this study fulfills function cascades under pathological state and shows great potential in IAI therapy.


Asunto(s)
Antibacterianos , Ácido Poliglutámico , Plata , Titanio , Animales , Titanio/química , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Ratones , Ácido Poliglutámico/química , Ácido Poliglutámico/análogos & derivados , Plata/química , Plata/farmacología , Propiedades de Superficie , Nanotubos/química , Células RAW 264.7 , Infecciones Relacionadas con Prótesis/tratamiento farmacológico , Oseointegración/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Cicatrización de Heridas/efectos de los fármacos , Prótesis e Implantes
4.
Biomaterials ; 313: 122774, 2025 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39208699

RESUMEN

Osteomyelitis (OM) is a progressive, inflammatory infection of bone caused predominately by Staphylococcus aureus. Herein, we engineered an antibiotic-eluting collagen-hydroxyapatite scaffold capable of eliminating infection and facilitating bone healing. An iterative freeze-drying and chemical crosslinking approach was leveraged to modify antibiotic release kinetics, resulting in a layered dual-release system whereby an initial rapid release of antibiotic to clear infection was followed by a sustained controlled release to prevent reoccurrence of infection. We observed that the presence of microbial collagenase accelerated antibiotic release from the crosslinked layer of the scaffold, indicating that the material is responsive to microbial activity. As exemplar drugs, vancomycin and gentamicin-eluting scaffolds were demonstrated to be bactericidal, and supported osteogenesis in vitro. In a pilot murine model of OM, vancomycin-eluting scaffolds were observed to reduce S. aureus infection within the tibia. Finally, in a rabbit model of chronic OM, gentamicin-eluting scaffolds both facilitated radial bone defect healing and eliminated S. aureus infection. These results show that antibiotic-eluting collagen-hydroxyapatite scaffolds are a one-stage therapy for OM, which when implanted into infected bone defects simultaneously eradicate infection and facilitate bone tissue healing.


Asunto(s)
Antibacterianos , Gentamicinas , Osteomielitis , Infecciones Estafilocócicas , Staphylococcus aureus , Andamios del Tejido , Animales , Andamios del Tejido/química , Antibacterianos/farmacología , Antibacterianos/química , Infecciones Estafilocócicas/tratamiento farmacológico , Osteomielitis/tratamiento farmacológico , Conejos , Staphylococcus aureus/efectos de los fármacos , Gentamicinas/farmacología , Gentamicinas/administración & dosificación , Gentamicinas/química , Gentamicinas/uso terapéutico , Ratones , Vancomicina/farmacología , Vancomicina/química , Vancomicina/administración & dosificación , Durapatita/química , Cinética , Cicatrización de Heridas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Colágeno/química , Femenino
5.
Biomaterials ; 313: 122800, 2025 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39241551

RESUMEN

The (002) crystallographic plane-oriented hydroxyapatite (HA) and anatase TiO2 enable favorable hydrophilicity, osteogenesis, and biocorrosion resistance. Thus, the crystallographic plane control in HA coating and crystalline phase control in TiO2 is vital to affect the surface and interface bioactivity and biocorrosion resistance of titanium (Ti) implants. However, a corresponding facile and efficient fabrication method is absent to realize the HA(002) mineralization and anatase TiO2 formation on Ti. Herein, we utilized the predominant Ti(0002) plane of the fibrous-grained titanium (FG Ti) to naturally form anatase TiO2 and further achieve a (002) basal plane oriented nanoHA (nHA) film through an in situ mild hydrothermal growth strategy. The formed FG Ti-nHA(002) remarkably improved hydrophilicity, mineralization, and biocorrosion resistance. Moreover, the nHA(002) film reserved the microgroove-like topological structure on FG Ti. It could enhance osteogenic differentiation through promoted contact guidance, showing one order of magnitude higher expression of osteogenic-related genes. On the other hand, the nHA(002) film restrained the osteoclast activity by blocking actin ring formation. Based on these capacities, FG Ti-nHA(002) improved new bone growth and binding strength in rabbit femur implantation, achieving satisfactory osseointegration within 2 weeks.


Asunto(s)
Durapatita , Oseointegración , Titanio , Titanio/química , Durapatita/química , Animales , Oseointegración/efectos de los fármacos , Conejos , Osteogénesis/efectos de los fármacos , Corrosión , Ratones , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Diferenciación Celular/efectos de los fármacos
6.
Biomaterials ; 312: 122714, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39079462

RESUMEN

Osteosarcoma, a malignant bone tumor often characterized by high hedgehog signaling activity, residual tumor cells, and substantial bone defects, poses significant challenges to both treatment response and postsurgical recovery. Here, we developed a nanocomposite hydrogel for the sustained co-delivery of bioactive magnesium ions, anti-PD-L1 antibody (αPD-L1), and hedgehog pathway antagonist vismodegib, to eradicate residual tumor cells while promoting bone regeneration post-surgery. In a mouse model of tibia osteosarcoma, this hydrogel-mediated combination therapy led to remarkable tumor growth inhibition and hence increased animal survival by enhancing the activity of tumor-suppressed CD8+ T cells. Meanwhile, the implanted hydrogel improved the microenvironment of osteogenesis through long-term sustained release of Mg2+, facilitating bone defect repair by upregulating the expression of osteogenic genes. After 21 days, the expression levels of ALP, COL1, RUNX2, and BGLAP in the Vis-αPD-L1-Gel group were approximately 4.1, 5.1, 5.5, and 3.4 times higher than those of the control, respectively. We believe that this hydrogel-based combination therapy offers a potentially valuable strategy for treating osteosarcoma and addressing the tumor-related complex bone diseases.


Asunto(s)
Neoplasias Óseas , Hidrogeles , Inmunoterapia , Nanocompuestos , Osteosarcoma , Osteosarcoma/patología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/terapia , Animales , Hidrogeles/química , Nanocompuestos/química , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Ratones , Inmunoterapia/métodos , Línea Celular Tumoral , Regeneración Ósea/efectos de los fármacos , Humanos , Osteogénesis/efectos de los fármacos , Antígeno B7-H1/metabolismo , Ratones Endogámicos BALB C , Magnesio/química
7.
Carbohydr Polym ; 346: 122666, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39245476

RESUMEN

The rising prevalence of bone injuries has increased the demand for minimally invasive treatments. Microbead hydrogels, renowned for cell encapsulation, provide a versatile substrate for bone tissue regeneration. They deliver bioactive agents, support cell growth, and promote osteogenesis, aiding bone repair and regeneration. In this study, we synthesized superparamagnetic iron oxide nanoparticles (Sp) coated with a calcium phosphate layer (m-Sp), achieving a distinctive flower-like micro-cluster morphology. Subsequently, sodium alginate (SA) microbead hydrogels containing m-Sp (McSa@m-Sp) were fabricated using a dropping gelation strategy. McSa@m-Sp is magnetically targetable, enhance cross-linking, control degradation rates, and provide strong antibacterial activity. Encapsulation studies with MC3T3-E1 cells revealed enhanced viability and proliferation. These studies also indicated significantly elevated alkaline phosphatase (ALP) activity and mineralization in MC3T3-E1 cells, as confirmed by Alizarin Red S (ARS) and Von Kossa staining, along with increased collagen production within the McSa@m-Sp microbead hydrogels. Immunocytochemistry (ICC) and gene expression studies supported the osteoinductive potential of McSa@m-Sp, showing increased expression of osteogenic markers including RUNX-2, collagen-I, osteopontin, and osteocalcin. Thus, McSa@m-Sp microbead hydrogels offer a promising strategy for multifunctional scaffolds in bone tissue engineering.


Asunto(s)
Alginatos , Regeneración Ósea , Fosfatos de Calcio , Proliferación Celular , Hidrogeles , Osteogénesis , Alginatos/química , Alginatos/farmacología , Animales , Ratones , Fosfatos de Calcio/química , Fosfatos de Calcio/farmacología , Osteogénesis/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Regeneración Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ingeniería de Tejidos/métodos , Línea Celular , Nanopartículas Magnéticas de Óxido de Hierro/química , Antibacterianos/farmacología , Antibacterianos/química
8.
Carbohydr Polym ; 346: 122614, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39245525

RESUMEN

Bone defects caused by trauma, infection and congenital diseases still face great challenges. Dihydromyricetin (DHM) is a kind of flavone extracted from Ampelopsis grossedentata, a traditional Chinese medicine. DHM can enhance the osteogenic differentiation of human bone marrow mesenchymal stem cells with the potential to promote bone regeneration. Hydrogel can be used as a carrier of DHM to promote bone regeneration due to its unique biochemical characteristics and three-dimensional structure. In this study, oxidized phellinus igniarius polysaccharides (OP) and L-arginine chitosan (CA) are used to develop hydrogel. The pore size and gel strength of the hydrogel can be changed by adjusting the oxidation degree of oxidized phellinus igniarius polysaccharides. The addition of DHM further reduce the pore size of the hydrogel (213 µm), increase the mechanical properties of the hydrogel, and increase the antioxidant and antibacterial activities of the hydrogel. The scavenging rate of DPPH are 72.30 ± 0.33 %, and the inhibition rate of E.coli and S.aureus are 93.12 ± 0.38 % and 94.49 ± 1.57 %, respectively. In addition, PCAD has good adhesion and biocompatibility, and its extract can effectively promote the osteogenic differentiation of MC3T3-E1 cells. Network pharmacology and molecular docking show that the promoting effect of DHM on osteogenesis may be achieved by activating the PI3K/AKT and MAPK signaling pathways. This is confirmed through in vitro cell experiments and in vivo animal experiments.


Asunto(s)
Regeneración Ósea , Quitosano , Flavonoles , Hidrogeles , Sistema de Señalización de MAP Quinasas , Osteogénesis , Fosfatidilinositol 3-Quinasas , Polisacáridos , Proteínas Proto-Oncogénicas c-akt , Quitosano/química , Quitosano/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Flavonoles/farmacología , Flavonoles/química , Ratones , Hidrogeles/química , Hidrogeles/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Polisacáridos/química , Polisacáridos/farmacología , Osteogénesis/efectos de los fármacos , Regeneración Ósea/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Arginina/química , Arginina/farmacología , Oxidación-Reducción/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/química , Escherichia coli/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Humanos , Antioxidantes/farmacología , Antioxidantes/química , Adhesivos/química , Adhesivos/farmacología
9.
Sci Rep ; 14(1): 20848, 2024 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242756

RESUMEN

The development of synthetic bone substitutes that equal or exceed the efficacy of autologous graft remains challenging. In this study, a rat calvarial defect model was used as a reference to investigate the influence of composition and architecture of 3D-printed cement, with or without bioactives, on tissue regeneration. Printable cement pastes were formulated by combining hyaluronic acid and cement precursors. Cementitious scaffolds were printed with 3 different patterns. After 7 weeks of implantation with or without bone marrow, multiparametric qualitative and quantitative assessments were performed using µCT, SEM, and histology. None of the set-up strategies was as efficient as autologous cancellous bone graft to repair calvarial defects. Nonetheless, the presence of scaffold improved the skull vault closure, particularly when the scaffold was soaked in total bone marrow before implantation. No significant effect of scaffold macro-architecture was observed on tissue mineralization. Magnesium phosphate-based scaffolds (MgP) seemed to induce higher bone formation than their calcium-phosphate-based counterparts. They also displayed a quicker biodegradation and sparse remaining material was found after 7 weeks of implantation. Although further improvements are required to reach clinical settings, this study demonstrated the potential of organo-mineral cements for bone regeneration and highlighted the peculiar properties of MgP-based cements.


Asunto(s)
Regeneración Ósea , Impresión Tridimensional , Cráneo , Andamios del Tejido , Animales , Regeneración Ósea/efectos de los fármacos , Andamios del Tejido/química , Ratas , Cráneo/efectos de los fármacos , Sustitutos de Huesos/química , Sustitutos de Huesos/farmacología , Fosfatos de Calcio/química , Fosfatos de Calcio/farmacología , Masculino , Cementos para Huesos/farmacología , Cementos para Huesos/química , Fosfatos/química , Osteogénesis/efectos de los fármacos , Microtomografía por Rayos X , Compuestos de Magnesio
10.
Bone Res ; 12(1): 52, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39231935

RESUMEN

Osteoporosis remains incurable. The most widely used antiresorptive agents, bisphosphonates (BPs), also inhibit bone formation, while the anabolic agent, teriparatide, does not inhibit bone resorption, and thus they have limited efficacy in preventing osteoporotic fractures and cause some side effects. Thus, there is an unmet need to develop dual antiresorptive and anabolic agents to prevent and treat osteoporosis. Hydroxychloroquine (HCQ), which is used to treat rheumatoid arthritis, prevents the lysosomal degradation of TNF receptor-associated factor 3 (TRAF3), an NF-κB adaptor protein that limits bone resorption and maintains bone formation. We attempted to covalently link HCQ to a hydroxyalklyl BP (HABP) with anticipated low antiresorptive activity, to target delivery of HCQ to bone to test if this targeting increases its efficacy to prevent TRAF3 degradation in the bone microenvironment and thus reduce bone resorption and increase bone formation, while reducing its systemic side effects. Unexpectedly, HABP-HCQ was found to exist as a salt in aqueous solution, composed of a protonated HCQ cation and a deprotonated HABP anion. Nevertheless, it inhibited osteoclastogenesis, stimulated osteoblast differentiation, and increased TRAF3 protein levels in vitro. HABP-HCQ significantly inhibited both osteoclast formation and bone marrow fibrosis in mice given multiple daily PTH injections. In contrast, HCQ inhibited marrow fibrosis, but not osteoclast formation, while the HABP alone inhibited osteoclast formation, but not fibrosis, in the mice. HABP-HCQ, but not HCQ, prevented trabecular bone loss following ovariectomy in mice and, importantly, increased bone volume in ovariectomized mice with established bone loss because HABP-HCQ increased bone formation and decreased bone resorption parameters simultaneously. In contrast, HCQ increased bone formation, but did not decrease bone resorption parameters, while HABP also restored the bone lost in ovariectomized mice, but it inhibited parameters of both bone resorption and formation. Our findings suggest that the combination of HABP and HCQ could have dual antiresorptive and anabolic effects to prevent and treat osteoporosis.


Asunto(s)
Conservadores de la Densidad Ósea , Resorción Ósea , Difosfonatos , Hidroxicloroquina , Ovariectomía , Animales , Ovariectomía/efectos adversos , Femenino , Ratones , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Difosfonatos/farmacología , Difosfonatos/uso terapéutico , Resorción Ósea/prevención & control , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Conservadores de la Densidad Ósea/farmacología , Conservadores de la Densidad Ósea/uso terapéutico , Ratones Endogámicos C57BL , Anabolizantes/farmacología , Anabolizantes/uso terapéutico , Osteogénesis/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Osteoporosis/prevención & control , Osteoporosis/metabolismo , Osteoporosis/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo
11.
J Cell Mol Med ; 28(17): e70040, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39219020

RESUMEN

Bone tissue engineering addresses the limitations of autologous resources and the risk of allograft disease transmission in bone diseases. In this regard, engineered three-dimensional (3D) models emerge as biomimetic alternatives to natural tissues, replicating intracellular communication. Moreover, the unique properties of super-paramagnetic iron oxide nanoparticles (SPIONs) were shown to promote bone regeneration via enhanced osteogenesis and angiogenesis in bone models. This study aimed to investigate the effects of SPION on both osteogenesis and angiogenesis and characterized a co-culture of Human umbilical vein endothelial cells (HUVEC) and MG-63 cells as a model of bone microtissue. HUVECs: MG-63s with a ratio of 4:1 demonstrated the best results among other cell ratios, and 50 µg/mL of SPION was the optimum concentration for maximum survival, cell migration and mineralization. In addition, the data from gene expression illustrated that the expression of osteogenesis-related genes, including osteopontin, osteocalcin, alkaline phosphatase, and collagen-I, as well as the expression of the angiogenesis-related marker, CD-31, and the tube formation, is significantly elevated when the 50 µg/mL concentration of SPION is applied to the microtissue samples. SPION application in a designed 3D bone microtissue model involving a co-culture of osteoblast and endothelial cells resulted in increased expression of specific markers related to angiogenesis and osteogenesis. This includes the design of a novel biomimetic model to boost blood compatibility and biocompatibility of primary materials while promoting osteogenic activity in microtissue bone models. Moreover, this can improve interaction with surrounding tissues and broaden the knowledge to promote superior-performance implants, preventing device failure.


Asunto(s)
Regeneración Ósea , Técnicas de Cocultivo , Células Endoteliales de la Vena Umbilical Humana , Osteogénesis , Ingeniería de Tejidos , Humanos , Regeneración Ósea/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Ingeniería de Tejidos/métodos , Nanopartículas de Magnetita/química , Neovascularización Fisiológica/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Nanopartículas Magnéticas de Óxido de Hierro/química , Supervivencia Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/citología
12.
Exp Clin Transplant ; 22(7): 551-558, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39223813

RESUMEN

OBJECTIVES: Mesenchymal stem cells are gaining attention in medicine because of their anti-inflammatory and immunosuppressive properties. Inflammatory conditions can modulate immune responses in mesenchymal stem cells.We investigated the expression of long noncoding RNAs (RMRP, MALT1, NKILA,THRIL, and Linc-MAF-4) in humanWharton jelly mesenchymal stem cells primed with polyinosinicpolycytidylic acid. MATERIALS AND METHODS: Mesenchymal stem cells were isolated from human Wharton jelly by the explant method. To determine the stem nature of the cells, we performed a differentiation test on bone and fat cells. We used flow cytometry analysis to determine surface markers. Umbilical cord mesenchymal stem cells (1 × 105) were cultured in T75 culture flasks in Dulbecco's modified Eagle medium containing 10% fetal bovine serum. After cells reached approximately 80% confluency, cells were exposed to 50 µg/mL of polyinosinic-polycytidylic acid, a Toll-like receptor 3 ligand, for 24, 48, and 72 hours. The control group were cells not exposed to polyinosinic-polycytidylic acid. Real-time polymerase chain reaction evaluated RMRP, MALAT1, NKILA, THRIL, and Linc-MAF-4 long noncoding RNAs. RESULTS: We observed significantly increased expression of NKILA inWharton jelly mesenchymal stem cells stimulated with polyinosinic-polycytidylic acid at 72 hours compared with expression level in the control group (P < .001). CONCLUSIONS: Results indicated that a potential mechanism by which the Toll-like receptor 3 ligand improves immunosuppression of mesenchymal stem cells can be attributed to the regulatory role of long noncoding RNAs, possibly through increased expression of anti-inflammatory long noncoding RNAs such as NKILA.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas , Poli I-C , ARN Largo no Codificante , Receptor Toll-Like 3 , Gelatina de Wharton , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 3/genética , Gelatina de Wharton/citología , Células Cultivadas , Poli I-C/farmacología , Diferenciación Celular/efectos de los fármacos , Factores de Tiempo , Regulación de la Expresión Génica , Osteogénesis/efectos de los fármacos
13.
Drug Des Devel Ther ; 18: 3903-3919, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39224902

RESUMEN

Purpose: Bone loss is a common complication of type 2 diabetes mellitus (T2DM). Circadian rhythms play a significant role in T2DM and bone remodeling. Eldecalcitol (ED-71), a novel active vitamin D analog, has shown promise in ameliorating T2DM. We aimed to investigate whether the circadian rhythm coregulator BMAL1 mediates the anti-osteoporotic effect of ED-71 in T2DM and its associated mechanisms. Methods: A T2DM mouse model was established using high-fat diet (HDF) and streptozotocin (STZ) injection, and blood glucose levels were monitored weekly. HE staining, Masson staining, and Micro-CT were performed to assess the changes in bone mass. IHC staining and IF staining were used to detect osteoblast status and BMAL1 expression and RT-qPCR was applied to detect the change of oxidative stress factors. In vitro, high glucose (HG) stimulation was used to simulate the cell environment in T2DM. RT-qPCR, Western blot, IF, ALP staining and AR staining were used to detect osteogenic differentiation and SIRT1/GSK3ß signaling pathway. DCFH-DA staining was used to detect reactive oxygen species (ROS) levels. Results: ED-71 increased bone mass and promoted osteogenesis in T2DM mice. Moreover, ED-71 inhibited oxidative stress and promoted BMAL1 expression in osteoblasts The addition of STL1267, an agonist of the BMAL1 transcriptional repressor protein REV-ERB, reversed the inhibitory effect of ED-71 on oxidative stress and the promotional effect on osteogenic differentiation. In addition, ED-71 facilitated SIRT1 expression and reduced GSK3ß activity. The inhibition of SIRT1 with EX527 partially attenuated ED-71's effects, whereas the GSK3ß inhibitor LiCl further enhanced ED-71's positive effects on BMAL1 expression. Conclusion: ED-71 ameliorates bone loss in T2DM by upregulating the circadian rhythm coregulator BMAL1 and promoting osteogenesis through inhibition of oxidative stress. The SIRT1/GSK3ß signaling pathway is involved in the regulation of BMAL1.


Asunto(s)
Factores de Transcripción ARNTL , Ritmo Circadiano , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Ratones Endogámicos C57BL , Osteogénesis , Regulación hacia Arriba , Animales , Factores de Transcripción ARNTL/metabolismo , Factores de Transcripción ARNTL/genética , Ratones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Osteogénesis/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Masculino , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Ritmo Circadiano/efectos de los fármacos , Estreptozocina , Vitamina D/farmacología , Vitamina D/análogos & derivados , Dieta Alta en Grasa , Células Cultivadas
14.
Redox Rep ; 29(1): 2398891, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39284587

RESUMEN

OBJECTIVE: The study will be to observe the effect of Sodium butyrate (NaB) on bone loss in lipopolysaccharide (LPS)-treated rats. METHODS: In the rat model, we observed that changes in the expression of oxidative stress regulators, inflammatory markers and target genes were measured by immunofluorescence and RT-PCR after treatment. Changes in viability and osteogenesis of MC3T3-E1, osteoclast differentiation in RAW264.7 cells in the presence of LPS were evaluated using CCK-8, ALP staining, RES staining, and TRAP staining. RESULTS: In vitro experiments have shown that LPS-induced inhibition of JC-1, SIRT1, GPX1 and SOD2 is associated with increased levels of inflammation and oxidative stress. In addition, NaB has been found to suppress oxidative stress, inflammation and Mito SOX, promote osteogenic differentiation, and inhibit osteoclast differentiation. In addition, NaB significantly promoted SITR1 expression, repaired impaired bone metabolism, and improved bone strength and bone mineral density. CONCLUSION: Given all this experimental evidence, the results strongly suggest that NaB can restore osteogenic activity in the presence of LPS by reducing intracellular ROS, inhibiting osteoclast differentiation and reducing bone loss in LPS-treated rat models.


Asunto(s)
Ácido Butírico , Inflamación , Lipopolisacáridos , Estrés Oxidativo , Animales , Lipopolisacáridos/toxicidad , Lipopolisacáridos/farmacología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ácido Butírico/farmacología , Inflamación/metabolismo , Inflamación/tratamiento farmacológico , Ratones , Células RAW 264.7 , Osteogénesis/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Diferenciación Celular/efectos de los fármacos , Densidad Ósea/efectos de los fármacos , Masculino , Ratas Sprague-Dawley , Huesos/efectos de los fármacos , Huesos/metabolismo
15.
Mol Med ; 30(1): 151, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39278948

RESUMEN

Erythropoietin (EPO), expressed in red blood progenitor cells, primarily regulates erythropoiesis by binding to its receptor. Besides anemia, recent studies have identified new therapeutic indications for EPO that are not connected to red blood cell formation. Elevated EPO levels harm bone homeostasis in adult organisms and are associated with increased osteoclast; however, the underlying molecular mechanisms remain unclear. This study demonstrated that EPO enhanced osteoclast differentiation and bone resorption in vitro. We showed that EPO promoted osteoclast formation by up-regulating PPARγ expression through activating the Jak2/ERK signaling pathway. Consistently, PPARγ antagonists rescued the hyperactivation of osteoclasts due to EPO, while PPARγ agonists reversed the EMP9-mediated decrease in osteoclast differentiation. Further, exposing female mice to EPO for two months led to a decrease in bone mass and increased osteoclast numbers. The present results suggested that EPO promotes osteoclastogenesis by regulating the Jak2/ERK/ PPARγ signaling pathway. From a clinical perspective, the risk of compromised bone health should be considered when using EPO to treat anemia in post-operative patients with intertrochanteric fractures of the femur, as it could significantly impact the patient's recovery and quality of life.


Asunto(s)
Diferenciación Celular , Eritropoyetina , Osteoclastos , PPAR gamma , Eritropoyetina/farmacología , Eritropoyetina/metabolismo , Animales , PPAR gamma/metabolismo , Osteoclastos/metabolismo , Osteoclastos/efectos de los fármacos , Ratones , Femenino , Diferenciación Celular/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Janus Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Humanos , Regulación hacia Arriba/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Resorción Ósea/metabolismo , Ratones Endogámicos C57BL
16.
Stem Cell Res Ther ; 15(1): 306, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39285500

RESUMEN

BACKGROUND: Influence on stem cells' angiogenesis and osteogenesis of NAD(P)H Quinone Dehydrogenase 1(NQO1) has been established, but its impact on dental pulp stem cells (DPSCs) is unexplored. An important strategy for the treatment of arteriosclerosis is to inhibit calcium deposition and to promote vascular repair and angiogenesis. This study investigated the function and mechanism of NQO1 on angiogenesis and osteogenesis of DPSCs, so as to provide a new ideal for the treatment of arteriosclerosis. METHODS: Co-culture of human DPSCs and human umbilical vein endothelial cells (HUVECs) was used to detect the angiogenesis ability. Alkaline phosphatase (ALP) activity, alizarin red staining (ARS), and transplantation of HA/tricalcium phosphate with DPSCs were used to detect osteogenesis. RESULTS: NQO1 suppressed in vitro tubule formation, migration, chemotaxis, and in vivo angiogenesis, as evidenced by reduced CD31 expression. It also enhanced ALP activity, ARS, DSPP expression and osteogenesis and boosted mitochondrial function in DPSCs. CoQ10, an electron transport chain activator, counteracted the effects of NQO1 knockdown on these processes. Additionally, NQO1 downregulated MAPK signaling, which was reversed by CoQ10 supplementation in DPSCs-NQO1sh. CONCLUSIONS: NQO1 inhibited angiogenesis and promoted the osteogenesis of DPSCs by suppressing MAPK signaling pathways and enhancing mitochondrial respiration.


Asunto(s)
Pulpa Dental , Células Endoteliales de la Vena Umbilical Humana , Sistema de Señalización de MAP Quinasas , NAD(P)H Deshidrogenasa (Quinona) , Neovascularización Fisiológica , Osteogénesis , Humanos , Osteogénesis/efectos de los fármacos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/genética , Neovascularización Fisiológica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Pulpa Dental/citología , Pulpa Dental/metabolismo , Técnicas de Cocultivo , Células Madre/metabolismo , Células Madre/citología , Células Cultivadas , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Ubiquinona/metabolismo , Animales , Diferenciación Celular , Angiogénesis
17.
J Orthop Surg Res ; 19(1): 531, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39218922

RESUMEN

BACKGROUND: Bone loss caused by microgravity exposure presents a serious threat to the health of astronauts, but existing treatment strategies have specific restrictions. This research aimed to investigate whether salidroside (SAL) can mitigate microgravity-induced bone loss and its underlying mechanism. METHODS: In this research, we used hindlimb unloading (HLU) and the Rotary Cell Culture System (RCCS) to imitate microgravity in vivo and in vitro. RESULTS: The results showed that salidroside primarily enhances bone density, microstructure, and biomechanical properties by stimulating bone formation and suppressing bone resorption, thereby preserving bone mass in HLU rats. In MC3T3-E1 cells cultured under simulated microgravity in rotary wall vessel bioreactors, the expression of osteogenic genes significantly increased after salidroside administration, indicating that salidroside can promote osteoblast differentiation under microgravity conditions. Furthermore, the Nrf2 inhibitor ML385 diminished the therapeutic impact of salidroside on microgravity-induced bone loss. Overall, this research provides the first evidence that salidroside can mitigate bone loss induced by microgravity exposure through stimulating the Nrf2/HO-1 pathway. CONCLUSION: These findings indicate that salidroside has great potential for treating space-related bone loss in astronauts and suggest that Nrf2/HO-1 is a viable target for counteracting microgravity-induced bone damage.


Asunto(s)
Glucósidos , Factor 2 Relacionado con NF-E2 , Fenoles , Simulación de Ingravidez , Glucósidos/farmacología , Glucósidos/uso terapéutico , Animales , Fenoles/farmacología , Fenoles/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , Ratones , Simulación de Ingravidez/efectos adversos , Ratas , Masculino , Hemo-Oxigenasa 1/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Ingravidez/efectos adversos , Osteogénesis/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Suspensión Trasera , Resorción Ósea/prevención & control , Resorción Ósea/etiología , Resorción Ósea/metabolismo , Densidad Ósea/efectos de los fármacos , Proteínas de la Membrana
18.
Int J Nanomedicine ; 19: 8751-8768, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39220194

RESUMEN

Purpose: Periodontitis is a chronic infectious disease characterized by progressive inflammation and alveolar bone loss. Forkhead box O1 (FoxO1), an important regulator, plays a crucial role in maintaining bone homeostasis and regulating macrophage energy metabolism and osteogenic differentiation of mesenchymal stem cells (MSCs). In this study, FoxO1 was overexpressed into small extracellular vesicles (sEV) using engineering technology, and effects of FoxO1-overexpressed sEV on periodontal tissue regeneration as well as the underlying mechanisms were investigated. Methods: Human periodontal ligament stem cell (hPDLSCs)-derived sEV (hPDLSCs-sEV) were isolated using ultracentrifugation. They were then characterized using transmission electron microscopy, Nanosight, and Western blotting analyses. hPDLSCs were treated with hPDLSCs-sEV in vitro after stimulation with lipopolysaccharide, and osteogenesis was evaluated. The effect of hPDLSCs-sEV on the polarization phenotype of THP-1 macrophages was also evaluated. In addition, we measured the reactive oxygen species (ROS) levels, adenosine triphosphate (ATP) production, mitochondrial characteristics, and metabolism of hPDLSCs and THP-1 cells. Experimental periodontitis was established in vivo in mice. HPDLSCs-sEV or phosphate-buffered saline (PBS) were injected into periodontal tissues for four weeks, and the maxillae were collected and assessed by micro-computed tomography, histological staining, and small animal in vivo imaging. Results: In vitro, FoxO1-overexpressed sEV promoted osteogenic differentiation of hPDLSCs in the inflammatory environment and polarized THP-1 cells from the M1 phenotype to the M2 phenotype. Furthermore, FoxO1-overexpressed sEV regulated the ROS level, ATP production, mitochondrial characteristics, and metabolism of hPDLSCs and THP-1 cells in the inflammatory environment. In the in vivo analyses, FoxO1-overexpressed sEV effectively promoted bone formation and inhibited inflammation. Conclusion: FoxO1-overexpressed sEV can regulate osteogenesis and immunomodulation. The ability of FoxO1-overexpressed sEV to regulate inflammation and osteogenesis can pave the way for the establishment of a therapeutic approach for periodontitis.


Asunto(s)
Vesículas Extracelulares , Proteína Forkhead Box O1 , Mitocondrias , Osteogénesis , Ligamento Periodontal , Periodontitis , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Osteogénesis/efectos de los fármacos , Animales , Humanos , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Mitocondrias/metabolismo , Periodontitis/terapia , Periodontitis/metabolismo , Ratones , Ligamento Periodontal/citología , Células THP-1 , Especies Reactivas de Oxígeno/metabolismo , Inflamación/metabolismo , Masculino , Diferenciación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Macrófagos/metabolismo , Regeneración , Células Cultivadas
19.
J Nanobiotechnology ; 22(1): 532, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39223550

RESUMEN

The recent development of nanobiomaterials has shed some light on the field of periodontal tissue regeneration. Laponite (LAP), an artificially synthesized two-dimensional (2D) disk-shaped nanosilicate, has garnered substantial attention in regenerative biomedical applications owing to its distinctive structure, exceptional biocompatibility and bioactivity. This study endeavors to comprehensively evaluate the influence of LAP on periodontal regeneration. The effects of LAP on periodontal ligament cells (PDLCs) on osteogenesis, cementogenesis and angiogenesis were systematically assessed, and the potential mechanism was explored through RNA sequencing. The results indicated that LAP improved osteogenic and cementogenic differentiation of PDLCs, the regulatory effects of LAP on PDLCs were closely correlated with activation of PI3K-AKT signaling pathway. Moreover, LAP enhanced angiogenesis indirectly via manipulating paracrine of PDLCs. Then, LAP was implanted into rat periodontal defect to confirm its regenerative potential. Both micro-CT and histological analysis indicated that LAP could facilitate periodontal tissue regeneration in vivo. These findings provide insights into the bioactivity and underlying mechanism of LAP on PDLCs, highlighting it might be a potential therapeutic option in periodontal therapy.


Asunto(s)
Diferenciación Celular , Osteogénesis , Ligamento Periodontal , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Ratas Sprague-Dawley , Regeneración , Transducción de Señal , Silicatos , Ligamento Periodontal/citología , Ligamento Periodontal/metabolismo , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Ratas , Osteogénesis/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Silicatos/farmacología , Silicatos/química , Humanos , Diferenciación Celular/efectos de los fármacos , Masculino , Células Cultivadas , Cementogénesis
20.
J Mater Sci Mater Med ; 35(1): 53, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39225913

RESUMEN

The interconnected structures in a 3D scaffold allows the movement of cells and nutrients. Therefore, this study aimed to investigate the in-vivo bioactivity of 3D-printed ß-tricalcium phosphate (ß-TCP) and hydroxyapatite (HAP) scaffolds that replicate biological bone. This study included 24-week-old male New Zealand white rabbits. A cylindrical bone defect with a diameter of 4.5 mm and a depth of 8 mm was created in the lateral aspect of the distal femur. A 3D-printed scaffold was implanted in the right femur (experimental side), whereas the left femur was kept free of implantation (control side). Micro-CT analysis and histological observations of the bone defect site were conducted at 4, 8, and 12 weeks postoperatively to track the bone repair progress. No evidence of new bone tissue formation was found in the medullary cavity of the bone defect on the control side. In contrast, on the experimental side, the 3D scaffold demonstrated sufficient bioactivity, leading to the growth of new bone tissue. Over time, new bone tissue gradually extended from the periphery toward the center, a phenomenon evident in both micro-CT images and biopsy staining. In the current study, we observed that the cells involved in bone metabolism adhered, spread, and proliferated on our newly designed 3D-printed scaffold with a bone microstructure. Therefore, it is suggested that this scaffold has sufficient bioactivity to induce new bone formation and could be expected to be a more useful artificial bone than the existing version.


Asunto(s)
Regeneración Ósea , Fosfatos de Calcio , Fémur , Impresión Tridimensional , Ingeniería de Tejidos , Andamios del Tejido , Microtomografía por Rayos X , Conejos , Animales , Fosfatos de Calcio/química , Andamios del Tejido/química , Masculino , Regeneración Ósea/efectos de los fármacos , Fémur/patología , Ingeniería de Tejidos/métodos , Osteogénesis/fisiología , Osteogénesis/efectos de los fármacos , Durapatita/química , Huesos/patología , Sustitutos de Huesos/química , Sustitutos de Huesos/farmacología , Ensayo de Materiales , Materiales Biocompatibles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA