Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.079
Filtrar
1.
J Transl Med ; 22(1): 437, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38720345

RESUMEN

BACKGROUND: Biological-derived hydroxyapatite is widely used as a bone substitute for addressing bone defects, but its limited osteoconductive properties necessitate further improvement. The osteo-immunomodulatory properties hold crucial promise in maintaining bone homeostasis, and precise modulation of macrophage polarization is essential in this process. Metabolism serves as a guiding force for immunity, and fluoride modification represents a promising strategy for modulating the osteoimmunological environment by regulating immunometabolism. In this context, we synthesized fluorinated porcine hydroxyapatite (FPHA), and has demonstrated its enhanced biological properties and osteogenic capacity. However, it remains unknown whether and how FPHA affects the immune microenvironment of the bone defects. METHODS: FPHA was synthesized and its composition and structural properties were confirmed. Macrophages were cultured with FPHA extract to investigate the effects of FPHA on their polarization and the related osteo-immune microenvironment. Furthermore, total RNA of these macrophages was extracted, and RNA-seq analysis was performed to explore the underlying mechanisms associated with the observed changes in macrophages. The metabolic states were evaluated with a Seahorse analyzer. Additionally, immunohistochemical staining was performed to evaluate the macrophages response after implantation of the novel bone substitutes in critical size calvarial defects in SD rats. RESULTS: The incorporation of fluoride ions in FPHA was validated. FPHA promoted macrophage proliferation and enhanced the expression of M2 markers while suppressing the expression of M1 markers. Additionally, FPHA inhibited the expression of inflammatory factors and upregulated the expression of osteogenic factors, thereby enhancing the osteogenic differentiation capacity of the rBMSCs. RNA-seq analysis suggested that the polarization-regulating function of FPHA may be related to changes in cellular metabolism. Further experiments confirmed that FPHA enhanced mitochondrial function and promoted the metabolic shift of macrophages from glycolysis to oxidative phosphorylation. Moreover, in vivo experiments validated the above results in the calvarial defect model in SD rats. CONCLUSION: In summary, our study reveals that FPHA induces a metabolic shift in macrophages from glycolysis to oxidative phosphorylation. This shift leads to an increased tendency toward M2 polarization in macrophages, consequently creating a favorable osteo-immune microenvironment. These findings provide valuable insights into the impact of incorporating an appropriate concentration of fluoride on immunometabolism and macrophage mitochondrial function, which have important implications for the development of fluoride-modified immunometabolism-based bone regenerative biomaterials and the clinical application of FPHA or other fluoride-containing materials.


Asunto(s)
Durapatita , Glucólisis , Macrófagos , Fosforilación Oxidativa , Ratas Sprague-Dawley , Animales , Durapatita/química , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Glucólisis/efectos de los fármacos , Ratas , Porcinos , Proliferación Celular/efectos de los fármacos , Masculino , Osteogénesis/efectos de los fármacos , Cráneo/patología , Cráneo/efectos de los fármacos , Ratones , Microambiente Celular/efectos de los fármacos , Células RAW 264.7 , Huesos/metabolismo , Huesos/efectos de los fármacos
2.
Nat Commun ; 15(1): 4160, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755128

RESUMEN

The regeneration of critical-size bone defects, especially those with irregular shapes, remains a clinical challenge. Various biomaterials have been developed to enhance bone regeneration, but the limitations on the shape-adaptive capacity, the complexity of clinical operation, and the unsatisfied osteogenic bioactivity have greatly restricted their clinical application. In this work, we construct a mechanically robust, tailorable and water-responsive shape-memory silk fibroin/magnesium (SF/MgO) composite scaffold, which is able to quickly match irregular defects by simple trimming, thus leading to good interface integration. We demonstrate that the SF/MgO scaffold exhibits excellent mechanical stability and structure retention during the degradative process with the potential for supporting ability in defective areas. This scaffold further promotes the proliferation, adhesion and migration of osteoblasts and the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in vitro. With suitable MgO content, the scaffold exhibits good histocompatibility, low foreign-body reactions (FBRs), significant ectopic mineralisation and angiogenesis. Skull defect experiments on male rats demonstrate that the cell-free SF/MgO scaffold markedly enhances bone regeneration of cranial defects. Taken together, the mechanically robust, personalised and bioactive scaffold with water-responsive shape-memory may be a promising biomaterial for clinical-size and irregular bone defect regeneration.


Asunto(s)
Materiales Biocompatibles , Regeneración Ósea , Fibroínas , Magnesio , Células Madre Mesenquimatosas , Osteogénesis , Andamios del Tejido , Fibroínas/química , Fibroínas/farmacología , Regeneración Ósea/efectos de los fármacos , Animales , Andamios del Tejido/química , Masculino , Osteogénesis/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Ratas , Magnesio/química , Magnesio/farmacología , Materiales Biocompatibles/química , Osteoblastos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ratas Sprague-Dawley , Agua/química , Proliferación Celular/efectos de los fármacos , Ingeniería de Tejidos/métodos , Cráneo/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Bombyx
3.
Biomed Mater ; 19(4)2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38688311

RESUMEN

This study investigated the effectiveness of bone regeneration upon the application of leptin and osteolectin to a three-dimensional (3D) printed poly(ϵ-caprolactone) (PCL) scaffold. A fused deposition modeling 3D bioprinter was used to fabricate scaffolds with a diameter of 4.5 mm, a height of 0.5 mm, and a pore size of 420-520 nm using PCL (molecular weight: 43 000). After amination of the scaffold surface for leptin and osteolectin adhesion, the experimental groups were divided into the PCL scaffold (control), the aminated PCL (PCL/Amine) scaffold, the leptin-coated PCL (PCL/Leptin) scaffold, and the osteolectin-coated PCL (PCL/Osteo) scaffold. Next, the water-soluble tetrazolium salt-1 (WST-1) assay was used to assess cell viability. All groups exhibited cell viability rates of >100%. Female 7-week-old Sprague-Dawley rats were used forin vivoexperiments. Calvarial defects were introduced on the rats' skulls using a 5.5 mm trephine bur. The rats were divided into the PCL (control), PCL/Leptin, and PCL/Osteo scaffold groups. The scaffolds were then inserted into the calvarial defect areas, and the rats were sacrificed after 8-weeks to analyze the defect area. Micro-CT analysis indicated that the leptin- and osteolectin-coated scaffolds exhibited significantly higher bone regeneration. Histological analysis revealed new bone and blood vessels in the calvarial defect area. These findings indicate that the 3D-printed PCL scaffold allows for patient-customized fabrication as well as the easy application of proteins like leptin and osteolectin. Moreover, leptin and osteolectin did not show cytotoxicity and exhibited higher bone regeneration potential than the existing scaffold.


Asunto(s)
Regeneración Ósea , Leptina , Poliésteres , Impresión Tridimensional , Ratas Sprague-Dawley , Andamios del Tejido , Leptina/metabolismo , Animales , Andamios del Tejido/química , Regeneración Ósea/efectos de los fármacos , Ratas , Poliésteres/química , Femenino , Ingeniería de Tejidos/métodos , Supervivencia Celular/efectos de los fármacos , Cráneo/efectos de los fármacos , Humanos , Osteogénesis/efectos de los fármacos , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Ensayo de Materiales
4.
Biomed Mater ; 19(3)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38626780

RESUMEN

Wool derived keratin, due to its demonstrated ability to promote bone formation, has been suggested as a potential bioactive material for implant surfaces. The aim of this study was to assess the effects of keratin-coated titanium on osteoblast functionin vitroand bone healingin vivo. Keratin-coated titanium surfaces were fabricated via solvent casting and molecular grafting. The effect of these surfaces on the attachment, osteogenic gene, and osteogenic protein expression of MG-63 osteoblast-like cells were quantifiedin vitro. The effect of these keratin-modified surfaces on bone healing over three weeks using an intraosseous calvaria defect was assessed in rodents. Keratin coating did not affect MG-63 proliferation or viability, but enhanced osteopontin, osteocalcin and bone morphogenetic expressionin vitro. Histological analysis of recovered calvaria specimens showed osseous defects covered with keratin-coated titanium had a higher percentage of new bone area two weeks after implantation compared to that in defects covered with titanium alone. The keratin-coated surfaces were biocompatible and stimulated osteogenic expression in adherent MG-63 osteoblasts. Furthermore, a pilot preclinical study in rodents suggested keratin may stimulate earlier intraosseous calvaria bone healing.


Asunto(s)
Regeneración Ósea , Proliferación Celular , Materiales Biocompatibles Revestidos , Queratinas , Osteoblastos , Osteogénesis , Cráneo , Titanio , Titanio/química , Osteoblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/metabolismo , Regeneración Ósea/efectos de los fármacos , Animales , Queratinas/química , Queratinas/metabolismo , Humanos , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Proliferación Celular/efectos de los fármacos , Cráneo/efectos de los fármacos , Cráneo/lesiones , Osteogénesis/efectos de los fármacos , Ratas , Propiedades de Superficie , Masculino , Línea Celular , Adhesión Celular/efectos de los fármacos , Ensayo de Materiales , Supervivencia Celular/efectos de los fármacos , Ratas Sprague-Dawley
5.
Carbohydr Polym ; 336: 122115, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38670750

RESUMEN

To alleviate skull defects and enhance the biological activity of taxifolin, this study utilized the thin-film dispersion method to prepare paclitaxel liposomes (TL). Thiolated chitosan (CSSH)-modified TL (CTL) was synthesized through charge interactions. Injectable hydrogels (BLG) were then prepared as hydrogel scaffolds loaded with TAX (TG), TL (TLG), and CTL (CTLG) using a Schiff base reaction involving oxidized dextran and carboxymethyl chitosan. The study investigated the bone reparative properties of CTLG through molecular docking, western blot techniques, and transcriptome analysis. The particle sizes of CTL were measured at 248.90 ± 14.03 nm, respectively, with zeta potentials of +36.68 ± 5.43 mV, respectively. CTLG showed excellent antioxidant capacity in vitro. It also has a good inhibitory effect on Escherichia coli and Staphylococcus aureus, with inhibition rates of 93.88 ± 1.59 % and 88.56 ± 2.83 % respectively. The results of 5-ethynyl-2 '-deoxyuridine staining, alkaline phosphatase staining and alizarin red staining showed that CTLG also had the potential to promote the proliferation and differentiation of mouse embryonic osteoblasts (MC3T3-E1). The study revealed that CTLG enhances the expression of osteogenic proteins by regulating the Wnt signaling pathway, shedding light on the potential application of TAX and bone regeneration mechanisms.


Asunto(s)
Proliferación Celular , Quitosano , Hidrogeles , Liposomas , Osteoblastos , Quercetina , Quercetina/análogos & derivados , Cráneo , Vía de Señalización Wnt , Animales , Quitosano/análogos & derivados , Quitosano/química , Quitosano/farmacología , Quercetina/farmacología , Quercetina/química , Liposomas/química , Vía de Señalización Wnt/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Proliferación Celular/efectos de los fármacos , Ratones , Cráneo/efectos de los fármacos , Cráneo/patología , Cráneo/metabolismo , Ratas , Regeneración Ósea/efectos de los fármacos , Ratas Sprague-Dawley , Osteogénesis/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Diferenciación Celular/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Masculino , Simulación del Acoplamiento Molecular
6.
Int J Biol Macromol ; 266(Pt 1): 130995, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38521323

RESUMEN

Critical-size bone defects are one of the main challenges in bone tissue regeneration that determines the need to use angiogenic and osteogenic agents. Rosuvastatin (RSV) is a class of cholesterol-lowering drugs with osteogenic potential. Magnesium oxide (MgO) is an angiogenesis component affecting apatite formation. This study aims to evaluate 3D-printed Polycaprolactone/ß-tricalcium phosphate/nano-hydroxyapatite/ MgO (PCL/ß-TCP/nHA/MgO) scaffolds as a carrier for MgO and RSV in bone regeneration. For this purpose, PCL/ß-TCP/nHA/MgO scaffolds were fabricated with a 3D-printing method and coated with gelatin and RSV. The biocompatibility and osteogenicity of scaffolds were examined with MTT, ALP, and Alizarin red staining. Finally, the scaffolds were implanted in a bone defect of rat's calvaria, and tissue regeneration was investigated after 3 months. Our results showed that the simultaneous presence of RSV and MgO improved biocompatibility, wettability, degradation rate, and ALP activity but decreased mechanical strength. PCL/ß-TCP/nHA/MgO/gelatin-RSV scaffolds produced sustained release of MgO and RSV within 30 days. CT images showed that PCL/ß-TCP/nHA/MgO/gelatin-RSV scaffolds filled approximately 86.83 + 4.9 % of the defects within 3 months and improved angiogenesis, woven bone, and osteogenic genes expression. These results indicate the potential of PCL/ß-TCP/nHA/MgO/gelatin-RSV scaffolds as a promising tool for bone regeneration and clinical trials.


Asunto(s)
Regeneración Ósea , Gelatina , Óxido de Magnesio , Osteogénesis , Impresión Tridimensional , Rosuvastatina Cálcica , Andamios del Tejido , Regeneración Ósea/efectos de los fármacos , Rosuvastatina Cálcica/farmacología , Rosuvastatina Cálcica/química , Andamios del Tejido/química , Gelatina/química , Animales , Ratas , Osteogénesis/efectos de los fármacos , Óxido de Magnesio/química , Óxido de Magnesio/farmacología , Poliésteres/química , Liberación de Fármacos , Durapatita/química , Durapatita/farmacología , Preparaciones de Acción Retardada/farmacología , Fosfatos de Calcio/química , Fosfatos de Calcio/farmacología , Cráneo/efectos de los fármacos , Ingeniería de Tejidos/métodos
7.
Adv Healthc Mater ; 13(12): e2303134, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38348511

RESUMEN

The effective repair of large bone defects remains a major challenge due to its limited self-healing capacity. Inspired by the structure and function of the natural periosteum, an electrospun biomimetic periosteum is constructed to programmatically promote bone regeneration using natural bone healing mechanisms. The biomimetic periosteum is composed of a bilayer with an asymmetric structure in which an aligned electrospun poly(ε-caprolactone)/gelatin/deferoxamine (PCL/GEL/DFO) layer mimics the outer fibrous layer of the periosteum, while a random coaxial electrospun PCL/GEL/aspirin (ASP) shell and PCL/silicon nanoparticles (SiNPs) core layer mimics the inner cambial layer. The bilayer controls the release of ASP, DFO, and SiNPs to precisely regulate the inflammatory, angiogenic, and osteogenic phases of bone repair. The random coaxial inner layer can effectively antioxidize, promoting cell recruitment, proliferation, differentiation, and mineralization, while the aligned outer layer can promote angiogenesis and prevent fibroblast infiltration. In particular, different stages of bone repair are modulated in a rat skull defect model to achieve faster and better bone regeneration. The proposed biomimetic periosteum is expected to be a promising candidate for bone defect healing.


Asunto(s)
Materiales Biomiméticos , Regeneración Ósea , Periostio , Poliésteres , Regeneración Ósea/efectos de los fármacos , Animales , Periostio/efectos de los fármacos , Ratas , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Poliésteres/química , Ratas Sprague-Dawley , Deferoxamina/farmacología , Deferoxamina/química , Gelatina/química , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Preparaciones de Acción Retardada/farmacocinética , Osteogénesis/efectos de los fármacos , Cráneo/efectos de los fármacos , Cráneo/lesiones , Masculino , Nanopartículas/química , Ingeniería de Tejidos/métodos , Diferenciación Celular/efectos de los fármacos , Andamios del Tejido/química
8.
Adv Healthc Mater ; 13(11): e2303851, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38226706

RESUMEN

Targeting macrophages can facilitate the site-specific repair of critical bone defects. Herein, a composite hydrogel, gelatin-Bletilla striata polysaccharide-mesoporous bioactive glass hydrogel (GBMgel), is constructed via the self-assembly of mesoporous bioactive glass on polysaccharide structures, through the Schiff base reaction. GBMgel can efficiently capture macrophages and drive the recruitment of seed stem cells and vascular budding required for regeneration in the early stages of bone injury, and the observed sustained release of inorganic silicon ions further enhances bone matrix deposition, mineralization, and vascular maturation. Moreover, the use of macrophage-depleted rat calvarial defect models further confirms that GBMgel, with ligand-selective macrophage targeting, increases the bone regeneration area and the proportion of mature bone. Mechanistic studies reveal that GBMgel upregulates the TLR4/NF-κB and MAPK macrophage pathways in the early stages and the JAK/STAT3 pathway in the later stages; thus initiating macrophage polarization at different time points. In conclusion, this study is based on the endogenous self-healing properties of bone macrophages, which enhances stem cell homing, and provides a research and theoretical basis upon which bone tissue can be reshaped and regenerated using the body's immune power, providing a new strategy for the treatment of critical bone defects.


Asunto(s)
Regeneración Ósea , Hidrogeles , Macrófagos , Animales , Regeneración Ósea/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratas , Ratas Sprague-Dawley , Ratones , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células RAW 264.7 , Ligandos , Masculino , Gelatina/química , Cráneo/efectos de los fármacos , Cráneo/patología , Cráneo/lesiones , Polisacáridos/química , Polisacáridos/farmacología
9.
Int J Mol Sci ; 22(22)2021 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-34830400

RESUMEN

This study aimed to evaluate the bone regeneration capacity of a customized alloplastic material and xenograft with recombinant human bone morphogenetic protein-2 (rhBMP-2). We prepared hydroxyapatite (HA)/tricalcium phosphate (TCP) pure ceramic bone blocks made using a 3D printing system and added rhBMP-2 to both materials. In eight beagle dogs, a total of 32 defects were created on the lower jaws. The defective sites of the negative control group were left untreated (N group; 8 defects), and those in the positive control group were filled with particle-type Bio-Oss (P group; 12 defects). The defect sites in the experimental group were filled with 3D-printed synthetic bone blocks (3D group; 12 defects). Radiographic and histological evaluations were performed after healing periods of 6 and 12 weeks and showed no significant difference in new bone formation and total bone between the P and 3D groups. The 3D-printed custom HA/TCP graft with rhBMP-2 showed bone regeneration effects similar to that of particulate Bio-Oss with rhBMP-2. Through further study and development, the application of 3D-printed customized alloplastic grafts will be extended to various fields of bone regeneration.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Regeneración Ósea/genética , Anomalías Maxilomandibulares/terapia , Desarrollo Maxilofacial/genética , Factor de Crecimiento Transformador beta/genética , Animales , Proteína Morfogenética Ósea 2/farmacología , Regeneración Ósea/efectos de los fármacos , Sustitutos de Huesos/farmacología , Fosfatos de Calcio/farmacología , Perros , Durapatita/farmacología , Humanos , Maxilares/patología , Anomalías Maxilomandibulares/genética , Anomalías Maxilomandibulares/patología , Impresión Tridimensional , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Cráneo/efectos de los fármacos , Cráneo/crecimiento & desarrollo , Cráneo/patología , Factor de Crecimiento Transformador beta/farmacología
10.
Cells ; 10(11)2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34831216

RESUMEN

Craniofacial bone defects can result from various disorders, including congenital malformations, tumor resection, infection, severe trauma, and accidents. Successfully regenerating cranial defects is an integral step to restore craniofacial function. However, challenges managing and controlling new bone tissue formation remain. Current advances in tissue engineering and regenerative medicine use innovative techniques to address these challenges. The use of biomaterials, stromal cells, and growth factors have demonstrated promising outcomes in vitro and in vivo. Natural and synthetic bone grafts combined with Mesenchymal Stromal Cells (MSCs) and growth factors have shown encouraging results in regenerating critical-size cranial defects. One of prevalent growth factors is Bone Morphogenetic Protein-2 (BMP-2). BMP-2 is defined as a gold standard growth factor that enhances new bone formation in vitro and in vivo. Recently, emerging evidence suggested that Megakaryocytes (MKs), induced by Thrombopoietin (TPO), show an increase in osteoblast proliferation in vitro and bone mass in vivo. Furthermore, a co-culture study shows mature MKs enhance MSC survival rate while maintaining their phenotype. Therefore, MKs can provide an insight as a potential therapy offering a safe and effective approach to regenerating critical-size cranial defects.


Asunto(s)
Cara/fisiología , Cráneo/fisiología , Ingeniería de Tejidos , Animales , Materiales Biocompatibles/farmacología , Regeneración Ósea/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Cráneo/efectos de los fármacos
11.
Int J Mol Sci ; 22(21)2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34768759

RESUMEN

Concentrated growth factor (CGF) is 100% blood-derived, cross-linked fibrin glue with platelets and growth factors. Human CGF clot is transformed into membrane by a compression device, which has been widely used clinically. However, the mechanical properties of the CGF membranes have not been well characterized. The aims of this study were to measure the tensile strength of human CGF membrane and observe its behavior as a scaffold of BMP-2 in ectopic site over the skull. The tensile test of the full length was performed at the speed of 2mm/min. The CGF membrane (5 × 5 × 2 mm3) or the CGF/BMP-2 (1.0 µg) membrane was grafted onto the skull periosteum of nude mice (5-week-old, male), and harvested at 14 days after the graft. The appearance and size of the CGF membranes were almost same for 7 days by soaking at 4 °C in saline. The average values of the tensile strength at 0 day and 7 days were 0.24 MPa and 0.26 MPa, respectively. No significant differences of both the tensile strength and the elastic modulus were found among 0, 1, 3, and 7 days. Supra-periosteal bone induction was found at 14 days in the CGF/BMP-2, while the CGF alone did not induce bone. These results demonstrated that human CGF membrane could become a short-term, sticky fibrin scaffold for BMP-2, and might be preserved as auto-membranes for wound protection after the surgery.


Asunto(s)
Proteína Morfogenética Ósea 2/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Periostio/efectos de los fármacos , Cráneo/efectos de los fármacos , Adulto , Animales , Proteína Morfogenética Ósea 2/uso terapéutico , Trasplante Óseo , Módulo de Elasticidad , Adhesivo de Tejido de Fibrina/química , Adhesivo de Tejido de Fibrina/farmacología , Adhesivo de Tejido de Fibrina/uso terapéutico , Voluntarios Sanos , Humanos , Péptidos y Proteínas de Señalización Intercelular/aislamiento & purificación , Masculino , Membranas/química , Membranas/metabolismo , Ratones Desnudos , Periostio/citología , Cráneo/citología , Resistencia a la Tracción , Cicatrización de Heridas/efectos de los fármacos
12.
Ann Biomed Eng ; 49(12): 3676-3684, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34608582

RESUMEN

Amelotin (AMTN) is a protein that is expressed during the maturation of dental enamel and has important role in enamel hydroxyapatite mineralization. However, it is not well understood whether AMTN has a strong mineral-promoting ability in bone. In this study, the effect of AMTN on bone healing was investigated using mice calvarial defect model in vivo, and the expression of bone marker genes and cell proliferation were investigated to clarify the role of AMTN in bone mineralization using mouse osteogenic cells (MC3T3-E1) in vitro. Collagen membranes, with or without recombinant human (rh) AMTN, were applied to calvarial defects created on the parietal bones of C57BL/6N mice. Microcomputed tomography and histological observation revealed that the defect largely filled with mineralized tissue by the rhAMTN-containing membrane in eight weeks. Moreover, CD31 positive cells were observed in the newly formed mineralized tissue and around the rhAMTN-containing membrane. In the presence of rhAMTN, the expression of the Spp1 gene in MC3T3-E1 cells significantly increased within ten days in an osteoinductive medium. Moreover, rhAMTN significantly enhanced MC3T3-E1 cell proliferation. These findings indicate that AMTN positively influences bone repair by promoting hydroxyapatite mineralization.


Asunto(s)
Proteínas del Esmalte Dental/farmacología , Cráneo/efectos de los fármacos , Cráneo/fisiopatología , Cicatrización de Heridas/efectos de los fármacos , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos BALB C , Osteoblastos/fisiología , Cráneo/diagnóstico por imagen , Cráneo/lesiones , Microtomografía por Rayos X
13.
J Mater Sci Mater Med ; 32(9): 125, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34524548

RESUMEN

The current study aimed to evaluate the effects of chemokine stromal cell-derived factor (SDF)-1α and platelet-rich plasma (PRP) on bone formation and angiogenesis, and to assess whether SDF-1α and PRP could function synergistically. Four evenly distributed defects (8 mm in diameter) were generated in the calvarial bones of New Zealand white rabbits. All rabbits received four treatment regimens containing autogenous bone particles (AB), SDF-1α, or PRP. AB group presented significantly less bone formation compared with the other three groups 2 and 4 weeks after surgery. The amount of newly formed bone in the AB+PRP+SDF-1α group was similar to that in the AB + SDF-1α group at the 4-week time-point (p = 0.65), and was much greater than that in the AB and AB+PRP group (p < 0.001). Meanwhile, more new blood vessels were formed in the AB+PRP, AB+SDF-1α, and AB+PRP+SDF-1α group versus the AB group. AB+PRP+SDF-1α group showed statistically increased angiogenesis compared with the AB+PRP and AB+SDF-1α groups (both p < 0.05) after treatment for 2 and 4 weeks. These findings indicated that SDF-1α and PRP might exhibit synergistic effects to promote angiogenesis in early bone regeneration.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Quimiocina CXCL12/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Plasma Rico en Plaquetas/fisiología , Cráneo/efectos de los fármacos , Animales , Trasplante Óseo/métodos , Osteogénesis/efectos de los fármacos , Conejos , Cráneo/irrigación sanguínea , Cráneo/patología , Cicatrización de Heridas/efectos de los fármacos
14.
Acta Biochim Biophys Sin (Shanghai) ; 53(11): 1516-1526, 2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34536273

RESUMEN

Vascularization is an important early indicator of osteogenesis involving biomaterials. Bone repair and new bone formation are associated with extensive neovascularization. Silicon-based biomaterials have attracted widespread attention due to their rapid vascularization. Although calcium phosphate cement (CPC) is a mature substitute for bone, the application of CPC is limited by its slow degradation and insufficient promotion of neovascularization. Calcium silicate (CS) has been shown to stimulate vascular endothelial proliferation. Thus, CS may be added to CPC (CPC-CS) to improve the biocompatibility and neovascularization of CPC. In the early phase of bone repair (the inflammatory phase), macrophages accumulate around the biomaterial and exert both anti- and pro-inflammatory effects. However, the effect of CPC-CS on macrophage polarization is not known, and it is not clear whether the effect on neovascularization is mediated through macrophage polarization. In the present study, we explored whether silicon-mediated macrophage polarization contributes to vascularization by evaluating the CPC-CS-mediated changes in the immuno-environment under different silicate ion contents both in vivo and in vitro. We found that the silicon released from CPC-CS can promote macrophage polarization into the M2 phenotype and rapid endothelial neovascularization during bone repair. Dramatic neovascularization and osteogenesis were observed in mouse calvarial bone defects implanted with CPC-CS containing 60% CS. These findings suggest that CPC-CS is a novel biomaterial that can modulate immune response, promote endothelial proliferation, and facilitate neovascularization and osteogenesis. Thus, CPC-CS shows potential as a bone substitute material.


Asunto(s)
Cementos para Huesos/farmacología , Regeneración Ósea/efectos de los fármacos , Compuestos de Calcio/farmacología , Fosfatos de Calcio/farmacología , Silicatos/farmacología , Silicio/farmacología , Cráneo/efectos de los fármacos , Animales , Cementos para Huesos/química , Compuestos de Calcio/química , Fosfatos de Calcio/química , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células RAW 264.7 , Silicatos/química , Silicio/química , Cráneo/irrigación sanguínea , Cráneo/lesiones
15.
Int Immunopharmacol ; 100: 108134, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34547679

RESUMEN

Transient receptor potential channel 6 (TRPC6) is a receptor-operated Ca2+ channel that plays an important role in Ca2+ influx in the majority of non-excitable cells and influences calcium signalling and cellular responses. Therefore, the purpose of the present study was to gain insight into the role of TRPC6 in the osteogenesis of periodontal ligament cells (PDLCs). By western blot and immunohistochemical staining, the protein level of TRPC6 was found to be increased in a time-dependent manner during osteoblastic differentiation of PDLCs. In addition, the TRPC6 inhibitor SKF96365 was used to block the function of TRPC6 and inhibit osteoblastic differentiation of PDLCs. The TRPC6 activator hyperforin dicyclohexylammonium salt (hyperforin DCHA) was used to activate TRPC6 and promote osteoblastic differentiation of PDLCs. In vivo, wild-type mice showed better bone regeneration than TRPC6-/- mice, suggesting that TRPC6 has notable osteogenic induction properties and is important for bone defect repair. In conclusion, the current data demonstrated that TRPC6 plays a significant role in osteoblastic differentiation of PDLCs, suggesting that it may be a promising therapeutic target in osteogenesis.


Asunto(s)
Osteoblastos/metabolismo , Osteogénesis , Ligamento Periodontal/metabolismo , Cráneo/metabolismo , Canal Catiónico TRPC6/metabolismo , Adolescente , Animales , Diferenciación Celular , Células Cultivadas , Niño , Femenino , Humanos , Imidazoles/farmacología , Masculino , Ratones Noqueados , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Ligamento Periodontal/efectos de los fármacos , Ligamento Periodontal/patología , Transducción de Señal , Cráneo/efectos de los fármacos , Cráneo/patología , Canal Catiónico TRPC6/efectos de los fármacos , Canal Catiónico TRPC6/genética , Terpenos/farmacología
16.
J Mater Sci Mater Med ; 32(9): 113, 2021 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-34453610

RESUMEN

Tissue regeneration and neovascularisation in cases of major bone loss is a challenge in maxillofacial surgery. The hypothesis of the present study is that the addition of resorbable bioactive ceramic Silica Calcium Phosphate Cement (SCPC) to Declluraized Muscle Scaffold (DSM) can expedite bone formation and maturation. Two surgical defect models were created in 18 nude transgenic mice. Group 1(n = 6), with a 2-mm decortication calvarial defect, was treated with a DSM/SCPC sheet over the corticated bone as an onlay then seeded with human Mesenchymal Stromal Cells hMSC in situ. In Group 2 (n = 6), a critical size (4 mm) calvarial defect was made and grafted with DSM/SCPC/in situ human bone marrow stromal cells (hMSCs). The control groups included Group 3 (n = 3) animals, with a 2-mm decortication defect treated with an onlay DSM sheet, and Group 4 (n = 3) animals, treated with critical size defect grafted with plain DSM. After 8 weeks, bone regeneration in various groups was evaluated using histology, immunohistochemistry and histomorphometry. New bone formation and maturation was superior in groups treated with DSM/SCPC/hMSC. The DMS/SCPC scaffold has the ability to augment and induce bone regeneration and neovascularisation in cases of major bone resorption and critical size defects.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Cerámica/uso terapéutico , Matriz Extracelular Descelularizada/uso terapéutico , Músculos/química , Implantación de Prótesis , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/uso terapéutico , Sustitutos de Huesos/química , Sustitutos de Huesos/uso terapéutico , Fosfatos de Calcio/química , Fosfatos de Calcio/farmacología , Células Cultivadas , Cerámica/química , Matriz Extracelular Descelularizada/química , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Desnudos , Ratones Transgénicos , Osteogénesis/efectos de los fármacos , Implantación de Prótesis/instrumentación , Implantación de Prótesis/métodos , Cráneo/efectos de los fármacos , Cráneo/patología , Cráneo/fisiopatología , Ingeniería de Tejidos/métodos
17.
J Mater Sci Mater Med ; 32(9): 109, 2021 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-34453621

RESUMEN

The aim of this study was to evaluate biocompatibility of hydroxyapatite (HAP) from fish waste using in vitro and in vivo assays. Fish samples (whitemouth croaker - Micropogonias furnieri) from the biowaste was used as HAP source. Pre-osteoblastic MC3T3-E1 cells were used in vitro study. In addition, bone defects were artificially created in rat calvaria and filled with HAP in vivo. The results demonstrated that HAP reduced cytotoxicity in pre-osteoblast cells after 3 and 6 days following HAP exposure. DNA concentration was lower in the HAP group after 6 days. Quantitative RT-PCR did not show any significant differences (p > 0.05) between groups. In vivo study revealed that bone defects filled with HAP pointed out moderate chronic inflammatory cells with slight proliferation of blood vessels after 7 and 15 days. Chronic inflammatory infiltrate was absent after 30 days of HAP exposure. There was also a decrease in the amount of biomaterial, being followed by newly formed bone tissue. All experimental groups also demonstrated strong RUNX-2 immoexpression in the granulation tissue as well as in cells in close contact with biomaterial. The number of osteoblasts inside the defect area was lower in the HAP group when compared to control group after 7 days post-implantation. Similarly, the osteoblast surface as well as the percentage of bone surface was higher in control group when compared with HAP group after 7 days post-implantation. Taken together, HAP from fish waste is a promising possibility that should be explored more carefully by tissue-engineering or biotechnology.


Asunto(s)
Durapatita/aislamiento & purificación , Durapatita/farmacología , Productos Pesqueros , Animales , Regeneración Ósea/efectos de los fármacos , Sustitutos de Huesos/química , Sustitutos de Huesos/aislamiento & purificación , Sustitutos de Huesos/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Productos Pesqueros/análisis , Ensayo de Materiales , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Osteogénesis/efectos de los fármacos , Perciformes , Ratas , Cráneo/efectos de los fármacos , Cráneo/fisiología , Residuos Sólidos/análisis
18.
ACS Appl Mater Interfaces ; 13(30): 35342-35355, 2021 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-34297530

RESUMEN

Growth-factor-free bone regeneration remains a challenge in craniofacial engineering. Here, we engineered an osteogenic niche composed of a commercially modified alginate hydrogel and whitlockite microparticles (WHMPs), which impart tunable physicochemical properties that can direct osteogenesis of human gingival mesenchymal stem cells (GMSCs). Our in vitro studies demonstrate that WHMPs induce osteogenesis of GMSCs more effectively than previously demonstrated hydroxyapatite microparticles (HApMPs). Alginate-WHMP hydrogels showed higher elasticity without any adverse effects on the viability of the encapsulated GMSCs. Moreover, the alginate-WHMP hydrogels upregulate the mitogen-activated protein kinase (MAPK) pathway, which in turn orchestrates several osteogenic markers, such as RUNX2 and OCN, in the encapsulated GMSCs. Concurrent coculture studies with human osteoclasts demonstrate that GMSCs encapsulated in alginate-WHMP hydrogels downregulate osteoclastic activity, potentially due to release of Mg2+ ions from the WHMPs along with secretion of osteoprotegerin from the GMSCs. In vivo studies demonstrated that the GMSCs encapsulated in our osteogenic niche were able to promote bone repair in calvarial defects in murine models. Altogether, our results confirmed the development of a promising treatment modality for craniofacial bone regeneration based on an injectable growth-factor-free hydrogel delivery system.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Fosfatos de Calcio/uso terapéutico , Hidrogeles/uso terapéutico , Cráneo/efectos de los fármacos , Alginatos/uso terapéutico , Animales , Diferenciación Celular/efectos de los fármacos , Células Inmovilizadas , Encía/citología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ratas Sprague-Dawley , Ingeniería de Tejidos/métodos
19.
ACS Appl Mater Interfaces ; 13(29): 33981-33994, 2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34260195

RESUMEN

Titanium (Ti) and titanium alloys have been widely used in the field of biomedicine. However, the unmatched biomechanics and poor bioactivities of conventional Ti implants usually lead to insufficient osseointegration. To tackle these challenges, it is critical to develop a novel Ti implant that meets the bioadaptive requirements for load-bearing critical bone defects. Notably, three-dimensional (3D)-printed Ti implants mimic the microstructure and mechanical properties of natural bones. Additionally, eco-friendly techniques based on inorganic-binding peptides have been applied to modify Ti surfaces. Herein, in our study, Ti surfaces were modified to reinforce osseointegration using chimeric peptides constructed by connecting W9, RP1P, and minTBP-1 directly or via (GP)4, respectively. PR1P is derived from the extracellular VEGF-binding domain of prominin-1, which increases the expression of VEGF and promotes the binding of VEGF to endothelial cells, thereby accelerating angiogenesis. W9 induces osteoblast differentiation in bone marrow mesenchymal stem cells and human mesenchymal stem cells to promote bone formation. Overall, chimeric peptides promote osseointegration by promoting angiogenesis and osteogenesis. Additionally, chimeric peptides with P3&4 were more effective than those with P1&2 in improving osseointegration, which might be ascribed to the capacity of P3&4 to provide a greater range for chimeric peptides to express their activity. This work successfully used chimeric peptides to modify 3D-Ti implant surfaces to improve osseointegration on the implant-bone surface.


Asunto(s)
Materiales Biocompatibles Revestidos/uso terapéutico , Oseointegración/efectos de los fármacos , Péptidos/uso terapéutico , Prótesis e Implantes , Secuencia de Aminoácidos , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Materiales Biocompatibles Revestidos/química , Módulo de Elasticidad , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Impresión Tridimensional , Ratas Sprague-Dawley , Cráneo/efectos de los fármacos , Propiedades de Superficie , Titanio/química
20.
PLoS One ; 16(7): e0254837, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34280226

RESUMEN

A firm connection of the bone-implant-fixation system is of utmost importance for patients with cranial defects. In order to improve the connection reliability, the current research focuses on finding the optimal fixation method, as well as selection of the implant manufacturing methods and the used materials. For the latter, implementation of bioactive materials such as hydroxyapatite or other calcium phosphates has also been considered in the literature. The aim of this study was to investigate the effect of gradual osseointegration on the biomechanical performance of cranial Ti6Al4V implants with a deposited HA coating as the osseointegration agent. This effect was assessed by two different computational approaches using finite element method (FEM) modeling. The values of key input parameters necessary for FEM were obtained from experimental plasma spray deposition of HA layers onto Ti6Al4V samples. Immediately upon implantation, the HA layer at the bone-implant contact area brought only a slight decrease in the values of von Mises stress in the implant and the micro-screws when compared to a non-coated counterpart; importantly, this was without any negative trade-off in other important characteristics. The major benefit of the HA coatings was manifested upon the modeled osseointegration: the results of both approaches confirmed a significant reduction of investigated parameters such as the total implant displacements (reduced from 0.050 mm to 0.012 mm and 0.002 mm while using Approach I and II, respectively) and stresses (reduced from 52 MPa to 10 MPa and 1 MPa) in the implanted components in comparison to non-coated variant. This is a very promising result for potential use of thermally sprayed HA coatings for cranial implants.


Asunto(s)
Materiales Biocompatibles Revestidos/farmacología , Implantes Dentales , Oseointegración/efectos de los fármacos , Aleaciones/química , Aleaciones/farmacología , Sustitutos de Huesos/química , Sustitutos de Huesos/farmacología , Fosfatos de Calcio/química , Fosfatos de Calcio/farmacología , Materiales Biocompatibles Revestidos/química , Durapatita/química , Durapatita/farmacología , Análisis de Elementos Finitos , Humanos , Ensayo de Materiales , Oseointegración/fisiología , Prótesis e Implantes , Cráneo/diagnóstico por imagen , Cráneo/efectos de los fármacos , Cráneo/patología , Titanio/química , Titanio/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...