Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 815
Filtrar
1.
Int J Nanomedicine ; 19: 3143-3166, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38585472

RESUMEN

Background: The ability of nanomaterials to induce osteogenic differentiation is limited, which seriously imped the repair of craniomaxillofacial bone defect. Magnetic graphene oxide (MGO) nanocomposites with the excellent physicochemical properties have great potential in bone tissue engineering. In this study, we aim to explore the craniomaxillofacial bone defect repairment effect of MGO nanocomposites and its underlying mechanism. Methods: The biocompatibility of MGO nanocomposites was verified by CCK8, live/dead staining and cytoskeleton staining. The function of MGO nanocomposites induced osteogenic differentiation of BMSCs was investigated by ALP activity detection, mineralized nodules staining, detection of osteogenic genes and proteins, and immune-histochemical staining. BMSCs with or without MGO osteogenic differentiation induction were collected and subjected to high-throughput circular ribonucleic acids (circRNAs) sequencing, and then crucial circRNA circAars was screened and identified. Bioinformatics analysis, Dual-luciferase reporter assay, RNA binding protein immunoprecipitation (RIP), fluorescence in situ hybridization (FISH) and osteogenic-related examinations were used to further explore the ability of circAars to participate in MGO nanocomposites regulation of osteogenic differentiation of BMSCs and its potential mechanism. Furthermore, critical-sized calvarial defects were constructed and were performed to verify the osteogenic differentiation induction effects and its potential mechanism induced by MGO nanocomposites. Results: We verify the good biocompatibility and osteogenic differentiation improvement effects of BMSCs mediated by MGO nanocomposites. Furthermore, a new circRNA-circAars, we find and identify, is obviously upregulated in BMSCs mediated by MGO nanocomposites. Silencing circAars could significantly decrease the osteogenic ability of MGO nanocomposites. The underlying mechanism involved circAars sponging miR-128-3p to regulate the expression of SMAD5, which played an important role in the repair craniomaxillofacial bone defects mediated by MGO nanocomposites. Conclusion: We found that MGO nanocomposites regulated osteogenic differentiation of BMSCs via the circAars/miR-128-3p/SMAD5 pathway, which provided a feasible and effective strategy for the treatment of craniomaxillofacial bone defects.


Asunto(s)
Grafito , MicroARNs , Nanocompuestos , MicroARNs/genética , Osteogénesis/genética , ARN Circular , Hibridación Fluorescente in Situ , Óxido de Magnesio , Células Cultivadas , Regeneración Ósea , Fenómenos Magnéticos , Diferenciación Celular
2.
J Ethnopharmacol ; 329: 118141, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38570149

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The active ingredients of traditional Chinese medicine (TCM), such as naringin (NG), Eucommiol, isopsoralen, icariin, Astragalus polysaccharides, and chondroitin sulfate, contained in Drynariae Rhizoma, Eucommiae Cortex, Psoralea corylifolia, Herba Epimedii, Astragalus radix and deer antler, are considered promising candidates for enhancing the healing of osteoporotic defects due to their outstanding bone homeostasis regulating properties. They are commonly used to activate bone repair scaffolds. AIM OF THE REVIEW: Bone repair scaffolds are inadequate to meet the demands of osteoporotic defect healing due to the lack of regulation of bone homeostasis. Therefore, selecting bone scaffolds activated with TCM to improve the therapeutic effect of repairing osteoporotic bone defects. MATERIALS AND METHODS: To gather information on bone scaffold activated by traditional Chinese medicine, we conducted a thorough search of several scientific databases, including Google Scholar, Web of Science, Scifinder, Baidu Scholar, PubMed, and China National Knowledge Infrastructure (CNKI). RESULTS: This review discusses the mechanism of TCM active ingredients in regulating bone homeostasis, including stimulating bone formation and inhibiting bone resorption process and the healing mechanism of traditional bone repair scaffolds activated by them for osteoporotic defect healing. CONCLUSION: In general, the introduction of TCM active ingredients provides a novel therapeutic approach for modulating bone homeostasis and facilitating osteoporotic defect healing, and also offers a new strategy for design of other unconventional bone defect healing materials.

3.
Biomater Adv ; 160: 213848, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38581745

RESUMEN

Tissue engineering shows promise in repairing extensive bone defects. The promotion of proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) by biological scaffolds has a significant impact on bone regeneration outcomes. In this study we used an injectable hydrogel, known as aminated mesoporous silica gel composite hydrogel (MSNs-NH2@GelMA), loaded with a natural drug, processed pyritum (PP), to promote healing of bone defects. The mechanical properties of the composite hydrogel were significantly superior to those of the blank hydrogel. In vitro experiments revealed that the composite hydrogel stimulated the osteogenic differentiation of BMSCs, and significantly increased the expression of type I collagen (Col 1), runt-related transcription factor 2 (Runx 2), alkaline phosphatase (ALP), osteocalcin (OCN). In vivo experiments showed that the composite hydrogel promoted the generation of new bones. These findings provide evidence that the composite hydrogel pyritum-loaded holds promise as a biomaterial for bone repair.

4.
ACS Appl Mater Interfaces ; 16(15): 18658-18670, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38587811

RESUMEN

Three-dimensional (3D)-printed biodegradable polymer scaffolds are at the forefront of personalized constructs for bone tissue engineering. However, it remains challenging to create a biological microenvironment for bone growth. Herein, we developed a novel yet feasible approach to facilitate biomimetic mineralization via self-adaptive nanotopography, which overcomes difficulties in the surface biofunctionalization of 3D-printed polycaprolactone (PCL) scaffolds. The building blocks of self-adaptive nanotopography were PCL lamellae that formed on the 3D-printed PCL scaffold via surface-directed epitaxial crystallization and acted as a linker to nucleate and generate hydroxyapatite crystals. Accordingly, a uniform and robust mineralized layer was immobilized throughout the scaffolds, which strongly bound to the strands and had no effect on the mechanical properties of the scaffolds. In vitro cell culture experiments revealed that the resulting scaffold was biocompatible and enhanced the proliferation and osteogenic differentiation of mouse embryolous osteoblast cells. Furthermore, we demonstrated that the resulting scaffold showed a strong capability to accelerate in vivo bone regeneration using a rabbit bone defect model. This study provides valuable opportunities to enhance the application of 3D-printed scaffolds in bone repair, paving the way for translation to other orthopedic implants.


Asunto(s)
Osteogénesis , Andamios del Tejido , Ratones , Animales , Conejos , Andamios del Tejido/química , Biomimética , Regeneración Ósea , Poliésteres/química , Ingeniería de Tejidos , Impresión Tridimensional
5.
Sci Bull (Beijing) ; 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38637224

RESUMEN

Orderly hierarchical structure with balanced mechanical, chemical, and electrical properties is the basis of the natural bone microenvironment. Inspired by nature, we developed a piezocatalytically-induced controlled mineralization strategy using piezoelectric polymer poly-L-lactic acid (PLLA) fibers with ordered micro-nano structures to prepare biomimetic tissue engineering scaffolds with a bone-like microenvironment (pcm-PLLA), in which PLLA-mediated piezoelectric catalysis promoted the in-situ polymerization of dopamine and subsequently regulated the controllable growth of hydroxyapatite crystals on the fiber surface. PLLA fibers, as analogs of mineralized collagen fibers, were arranged in an oriented manner, and ultimately formed a bone-like interconnected pore structure; in addition, they also provided bone-like piezoelectric properties. The uniformly sized HA nanocrystals formed by controlled mineralization provided a bone-like mechanical strength and chemical environment. The pcm-PLLA scaffold could rapidly recruit endogenous stem cells, and promote their osteogenic differentiation by activating cell membrane calcium channels and PI3K signaling pathways through ultrasound-responsive piezoelectric signals. In addition, the scaffold also provided a suitable microenvironment to promote macrophage M2 polarization and angiogenesis, thereby enhancing bone regeneration in skull defects of rats. The proposed piezocatalytically-induced controllable mineralization strategy provides a new idea for the development of tissue engineering scaffolds that can be implemented for multimodal physical stimulation therapy.

6.
Biomater Adv ; 160: 213856, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38640877

RESUMEN

Large bone defects cause significant clinical challenges due to the lack of optimal grafts for effective regeneration. The tissue engineering way that requires the combination of biomaterials scaffold, stem cells and proper bioactive factors is a prospective method for large bone repair. Here, we synthesized a three-arm host-guest supramolecule (HGSM) to covalently crosslinking with the naturally derived polymer methacrylated silk fibroin (SFMA). The combination of HGSM and SFMA can form a high strength double-crosslinked hydrogel HGSFMA, that serve as the hydrogel scaffold for bone marrow mesenchymal stem cells (BMSCs) growing. Icariin (ICA) loaded in the HGSFMA hydrogel can promote the osteogenesis efficiency of BMSCs and inhibit the osteoclasts differentiation. Our findings demonstrated that the HGSFMA/ICA hydrogel effectively promoted the in vitro adhesion, proliferation, and osteogenic differentiation of BMSCs. Rat femoral defects model show that this hydrogel can completely repair femoral damage within 4 weeks and significantly promote the secretion of osteogenesis-related proteins. In summary, we have prepared an effective biomimetic bone carrier, offering a novel strategy for bone regeneration and the treatment of large-scale bone defects.

7.
Small ; : e2311344, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38661278

RESUMEN

The effect of immunoinflammation on bone repair during the recovery process of bone defects needs to be further explored. It is reported that Mg2+ can promote bone repair with immunoregulatory effect, but the underlying mechanism on adaptive immunity is still unclear. Here, by using chitosan and hyaluronic acid-coated Mg2+ (CSHA-Mg) in bone-deficient mice, it is shown that Mg2+ can inhibit the activation of CD4+ T cells and increase regulatory T cell formation by inducing immunosuppressive dendritic cells (imDCs). Mechanistically, Mg2+ initiates the activation of the MAPK signaling pathway through TRPM7 channels on DCs. This process subsequently induces the downstream HIF-1α expression, a transcription factor that amplifies TGF-ß production and inhibits the effective T cell function. In vivo, knock-out of HIF-1α in DCs or using a HIF-1α inhibitor PX-478 reverses inhibition of bone inflammation and repair promotion upon Mg2+-treatment. Moreover, roxadustat, which stabilizes HIF-1α protein expression, can significantly promote immunosuppression and bone repair in synergism with CSHA-Mg. Thus, the findings identify a key mechanism for DCs and its HIF-1α-TGF-ß axis in the induction of immunosuppressive bone microenvironment, providing potential targets for bone regeneration.

9.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(1): 28-36, 2024 Feb 01.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38475948

RESUMEN

OBJECTIVES: This study aims to compare the osteogenic effects of implanting demineralized dentin matrix and acellular dentin matrix in bone defect areas. METHODS: Demineralized dentin matrix and acellular dentin matrix were prepared. Twenty-four male SPF-grade SD rats were randomly divided into four groups: demineralized group (group A), acelluar group (group B), Bio-Oss bone powder group (group C), and blank control group (group D), with six rats in each group. All rats were subjected to general anesthesia to prepare bilateral femoral bone defects. Rats in groups A, B, and C were implanted with demineralized dentin matrix, acellular dentin matrix, and Bio-Oss bone powder at the bone defect area, respectively, while rats in group D were not implanted with any material. At 4 and 8 weeks after surgery, three rats were randomly executed in each group. The healing of the bone defect area was analyzed through gross observation. The concentrations of osteogenic indicators bone morphogenetic protein-2 (BMP-2) and alkaline phosphatase (ALP) were detected by serology. The distribution of high-density gray area (representing bone healing) in the bone defect area was observed by X-ray examination, and the formation of new bone was observed by histomorphology. The rate of new bone formation was calculated. RESULTS: At 4 and 8 weeks, the bone formation ability of group A was more active than that of the other groups; the concentrations of BMP-2 and ALP in group A were higher than those in the other groups, and the differences were statistically significant (P<0.05). At 8 weeks, the imaging observation showed that the high-density gray area in the bone defect of group A was evenly distributed. The histomorphological observation showed the regular arrangement of bone matrix in group A. The rate of new bone formation in group A was 28.51%±0.55% at 4 weeks and 32.57%±2.28% at 8 weeks, both of which were significantly higher than those in the other groups (P<0.05). CONCLUSIONS: Demineralized dentin matrix has better osteogenic potential than acellular dentin matrix.


Asunto(s)
Dentina , Minerales , Osteogénesis , Ratas , Masculino , Animales , Ratas Sprague-Dawley , Polvos
10.
Biomolecules ; 14(3)2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38540737

RESUMEN

Bone morphogenetic protein (BMP) and platelet-derived growth factor (PDGF) are known to regulate/stimulate osteogenesis, playing vital roles in bone homeostasis, rendering them strong candidates for osteoporosis treatment. We evaluated the effects of recombinant human BMP-7 (rhBMP7) and PDGF-BB (rhPDGF-BB) in an oophorectomy-induced osteoporosis rat model. Forty Sprague Dawley rats underwent oophorectomy surgery; treatments commenced on the 100th day post-surgery when all animals exhibited signs of osteoporosis. These peptide growth factors were administered intraocularly (iv) once or twice a week and the animals were monitored for a total of five weeks. Two weeks after the conclusion of the treatments, the animals were euthanized and tissues were collected for assessment of alkaline phosphatase, X-ray, micro-CT, and histology. The results indicate that the most promising treatments were 20 µg/kg rhPDGF-BB + 30 µg/kg rhBMP-7 twice a week and 30 µg/kg BMP-7 twice a week, showing significant increases of 15% (p < 0.05) and 13% (p < 0.05) in bone volume fraction and 21% (p < 0.05) and 23% (p < 0.05) in trabecular number, respectively. In conclusion, rhPDGF-BB and rhBMP-7 have demonstrated the ability to increase bone volume and density in this osteoporotic animal model, establishing them as potential candidates for osteoporosis treatment.


Asunto(s)
Proteína Morfogenética Ósea 7 , Osteoporosis , Humanos , Ratas , Animales , Becaplermina/farmacología , Proteínas Proto-Oncogénicas c-sis/farmacología , Proteínas Proto-Oncogénicas c-sis/uso terapéutico , Proteína Morfogenética Ósea 7/farmacología , Proteína Morfogenética Ósea 7/uso terapéutico , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Proteínas Morfogenéticas Óseas , Osteoporosis/tratamiento farmacológico , Proteína Morfogenética Ósea 2
11.
Int J Mol Sci ; 25(6)2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38542453

RESUMEN

Promoting the efficiency of bone regeneration in bone loss diseases is a significant clinical challenge. Traditional therapies often fail to achieve better therapeutic outcomes and shorter treatment times. However, in recent years, extracellular vesicles (EVs) have gained significant attention due to their exceptional osteogenic function in bone regeneration and superior therapeutic effects compared to traditional cell therapy. EVs have emerged as a promising therapy for tissue defect regeneration due to their various physiological functions, such as regulating the immune response and promoting tissue repair and regeneration. Moreover, EVs have good biocompatibility, low immunogenicity, and long-term stability, and can be improved through pretreatment and other methods. Studies investigating the mechanisms by which extracellular vesicles promote bone regeneration and applying EVs from different sources using various methods to animal models of bone defects have increased. Therefore, this paper reviews the types of EVs used for bone regeneration, their sources, roles, delivery pathways, scaffold biomaterials, and applications.


Asunto(s)
Enfermedades Óseas , Vesículas Extracelulares , Animales , Regeneración Ósea/fisiología , Osteogénesis , Vesículas Extracelulares/metabolismo , Materiales Biocompatibles/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos , Enfermedades Óseas/terapia , Enfermedades Óseas/metabolismo
12.
ACS Biomater Sci Eng ; 10(4): 2062-2067, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38466032

RESUMEN

Brushite calcium phosphate cement (brushite CPC) is a prospective bone repair material due to its ideal resorption rates in vivo. However, the undesirable mechanical property and bioactivity limited its availability in clinic application. To address this issue, incorporating polymeric additives has emerged as a viable solution. In this study, poly(ethylene glycol) dicarboxylic acid, PEG(COOH), was synthesized and employed as the polymeric additive. The setting behavior, anti-washout ability, mechanical property, degradation rate, and osteogenic capacity of brushite CPC were regulated by incorporating PEG(COOH). The incorporation of PEG(COOH) with carboxylic acid groups demonstrated a positive effect on both mechanical properties and osteogenic activity in bone repair. This study offers valuable insights and suggests a promising strategy for the development of materials in bone tissue engineering.


Asunto(s)
Cementos para Huesos , Polietilenglicoles , Polietilenglicoles/farmacología , Estudios Prospectivos , Cementos para Huesos/farmacología , Fosfatos de Calcio/farmacología , Polímeros , Ácidos Dicarboxílicos/farmacología
13.
ACS Biomater Sci Eng ; 10(4): 2385-2397, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38538611

RESUMEN

Bone is a complex organic-inorganic composite tissue composed of ∼30% organics and ∼70% hydroxyapatite (HAp). Inspired by this, we used 30% collagen and 70% HAp extracted from natural bone using the calcination method to generate a biomimetic bone composite hydrogel scaffold (BBCHS). In one respect, BBCHS, with a fixed proportion of inorganic and organic components similar to natural bone, exhibits good physical properties. In another respect, the highly biologically active and biocompatible HAp from natural bone effectively promotes osteogenic differentiation, and type I collagen facilitates cell adhesion and spreading. Additionally, the well-structured porosity of the BBCHS provides sufficient growth space for bone marrow mesenchymal stem cells (BMSCs) while promoting substance exchange. Compared to the control group, the new bone surface of the defective location in the B-HA70+Col group is increased by 3.4-fold after 8 weeks of in vivo experiments. This strategy enables the BBCHS to closely imitate the chemical makeup and physical structure of natural bone. With its robust biocompatibility and osteogenic activity, the BBCHS can be easily adapted for a wide range of bone repair applications and offers promising potential for future research and development.


Asunto(s)
Durapatita , Osteogénesis , Durapatita/farmacología , Durapatita/química , Andamios del Tejido/química , Biomimética , Hidrogeles/farmacología , Colágeno/farmacología
14.
Curr Med Chem ; 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38523515

RESUMEN

Since hydroxyapatite (HAp) is an important constituent of bone and teeth, it has excellent biocompatibility and bioactivity, good osteoconductive effects and the ability to induce bone formation as a material for bone or tooth repair and replacement. At present, widely used HAp microspheres have some characteristics, such as large specific surface area, light mass, good injection properties, good fluidity, and low aggregation ability, but they are difficult to really meet the biological and clinical needs due to their own mechanical property defects, such as low strength, brittleness, and poor plasticity. Based on the current research status of HAp microspheres, we summarize the research progress of various types of composite microspheres, including inorganic materials, natural polymer materials and synthetic polymer materials, and further analyze the advantages of HAp composite microspheres loaded with drug molecules, proteins and bioactive factors, so as to explore the development prospect of HAp composite microspheres as scaffolds for constructing sustained release systems. It provides a theoretical basis and research direction to prepare HAp composite micro-spheres with superior comprehensive properties so that they can be better applied in bone tissue regeneration and tooth regeneration engineering.

15.
ACS Biomater Sci Eng ; 10(4): 2414-2425, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38446137

RESUMEN

Bone defects are a common and challenging orthopedic problem with poor self-healing ability and long treatment cycles. The difficult-to-heal bone defects cause a significant burden of medical expenses on patients. Currently, biomaterials with mechanical stability, long-lasting action, and osteogenic activity are considered as a suitable way to effectively heal bone defects. Here, an injectable double network (DN) hydrogel prepared using physical and chemical cross-linking methods is designed. The first rigid network is constructed using methylpropenylated hyaluronic acid (HAMA), while the addition of chitosan oligosaccharide (COS) forms a second flexible network by physical cross-linking. The mesoporous silica nanoparticles (MSN) loaded with bone morphogenetic protein-4 (BMP-4) were embedded into DN hydrogel, which not only enhanced the mechanical stability of the hydrogel, but also slowly released BMP-4 to achieve long-term skull repair. The designed composite hydrogel showed an excellent compression property and deformation resistance. In vitro studies confirmed that the HAMA/COS/MSN@BMP-4 hydrogel had good biocompatibility and showed great potential in supporting proliferation and osteogenic differentiation of mouse embryo osteoblast precursor (MC3T3-E1) cells. Furthermore, in vivo studies confirmed that the DN hydrogel successfully filled and closed irregular skull defect wounds, effectively promoted bone regeneration, and significantly promoted bone repair compared with the control group. In addition, HAMA/COS/MSN@BMP-4 hydrogel precursor solution can quickly form hydrogel in situ at the wound by ultraviolet light, which can be applied to the closure and repair of wounds of different shapes, which provides the new way for the treatment of bone defects.


Asunto(s)
Hidrogeles , Nanopartículas , Ratones , Animales , Humanos , Hidrogeles/farmacología , Hidrogeles/química , Osteogénesis , Dióxido de Silicio/farmacología , Proteína Morfogenética Ósea 2/química , Proteína Morfogenética Ósea 2/farmacología , Cráneo/cirugía , Cráneo/lesiones , Nanopartículas/química
16.
Prog Biophys Mol Biol ; 188: 55-67, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38493961

RESUMEN

Bone repair is faced with obstacles such as slow repair rates and limited bone regeneration capacity. Delayed healing even nonunion could occur in bone defects, influencing the life quality of patients severely. Photobiomodulation (PBM) utilizes different light sources to derive beneficial therapeutic effects with the advantage of being non-invasive and painless, providing a promising strategy for accelerating bone repair. In this review, we summarize the parameters, mechanisms, and effects of PBM regulating bone repair, and further conclude the current clinical application of PBM devices in bone repair. The wavelength of 635-980 nm, the output power of 40-100 mW, and the energy density of less than 100 J/cm2 are the most commonly used parameters. New technologies, including needle systems and biocompatible and implantable optical fibers, offer references to realize an efficient and safe strategy for bone repair. Further research is required to establish the reliability of outcomes from in vivo and in vitro studies and to standardize clinical trial protocols.


Asunto(s)
Terapia por Luz de Baja Intensidad , Humanos , Terapia por Luz de Baja Intensidad/métodos , Reproducibilidad de los Resultados
17.
Front Bioeng Biotechnol ; 12: 1339135, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38476968

RESUMEN

Introduction: Bone tissue engineering is considered the ideal approach for bone repair. Mesoporous bioactive glass (MBG) possesses the characteristics of high drug-loading capacity and bioactivity. Low-intensity pulsed ultrasound contributes to promoting fracture healing and bone defect repair, and dimethyloxalyl glycine (DMOG) is a small molecular inhibitor that can suppress prolyl hydroxylase, reducing the degradation of hypoxia-inducible factor. Methods: In this study, we proposed to prepare DMOG-loaded MBG/poly(D,L-lactide) composite scaffolds (DMOG-MBG/PDLLA) for promoting bone repair. The effects of ultrasound stimulation and DMOG release on the cell responses of rat bone marrow mesenchymal stem cells (BMSCs) and human umbilical vein endothelial cells (HUVECs) and bone repair in vivo were investigated. Results and Discussion: The results showed that both ultrasound stimulation and DMOG release could promote the proliferation, adhesion and differentiation of BMSCs and HUVECs, respectively. After the implantation of scaffolds in rat cranial bone defect model for 8 weeks, the results indicated that the combined ultrasound stimulation and DMOG release contributed to the highest ability for promoting bone repair. Hence, the DMOG-MBG/PDLLA scaffolds with ultrasound stimulation are promising for application in bone repair.

18.
Biomedicines ; 12(2)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38397946

RESUMEN

In orthopedics, the repair of bone defects remains challenging. In previous research reports, magnesium phosphate cements (MPCs) were widely used because of their excellent mechanical properties, which have been widely used in the field of orthopedic medicine. We built a new k-struvite (MPC) cement obtained from zinc oxide (ZnO) and assessed its osteogenic properties. Zinc-doped magnesium phosphate cement (ZMPC) is a novel material with good biocompatibility and degradability. This article summarizes the preparation method, physicochemical properties, and biological properties of ZMPC through research on this material. The results show that ZMPC has the same strength and toughness (25.3 ± 1.73 MPa to 20.18 ± 2.11 MPa), that meet the requirements of bone repair. Furthermore, the material can gradually degrade (12.27% ± 1.11% in 28 days) and promote osteogenic differentiation (relative protein expression level increased 2-3 times) of rat bone marrow mesenchymal stem cells (rBMSCs) in vitro. In addition, in vivo confirmation revealed increased bone regeneration in a rat calvarial defect model compared with MPC alone. Therefore, ZMPC has broad application prospects and is expected to be an important repair material in the field of orthopedic medicine.

19.
Adv Healthc Mater ; : e2304585, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38411324

RESUMEN

The innate immune response is crucial to inflammation, but how neutrophils and macrophages act in bone repair and tissue engineering treatment strategies await clarification. In this study, it is found that N2 neutrophils release stronger "eat me" signals to induce macrophage phagocytosis and polarize into the M2 anti-inflammatory phenotype. Guided by this biological mechanism, a mesoporous bioactive glass scaffold (MBG) is filled with hyaluronic acid methacryloyl (HAMA) hydrogel loaded with Transforming growth factor-ß1 (TGFß1) adenovirus (Ad@H), constructing a genetically engineered composite scaffold (Ad@H/M). The scaffold not only has good hydrophilicity and biocompatibility, but also provides mechanical stress support for bone repair. Adenovirus infection quickly induces N2 neutrophils, upregulating NF-κB and MAPK signaling pathways through Toll-like receptor 4 (TLR4) to promote the inflammatory response and macrophage phagocytosis. Macrophages perform phagocytosis and polarize towards the M2 phenotype, mediating the inflammatory response by inhibiting the PI3K-AKT-NF-κB pathway, maintaining homeostasis of the osteogenic microenvironment. The role of the Ad@H/M scaffold in regulating early inflammation and promoting long-term bone regeneration is further validated in vivo. In brief, this study focuses on the cascade of reactions between neutrophils and macrophage subtypes, and reports a composite scaffold that coordinates the innate immune response to promote bone repair.

20.
Int J Biol Macromol ; 263(Pt 2): 130368, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38401584

RESUMEN

Surgical resection remains the primary treatment modality for bone tumors. However, it is prone to local bone defects and tumor recurrence. Therefore, there is an urgent need for multifunctional biomaterials that combine tumor treatment and bone repair after bone tumor surgery. Herein, a chitosan composite scaffold (CS/DOX@Ti-MOF) was designed for both tumor therapy and bone repair. Among them, the amino-functionalized Ti-based metal-organic framework (NH2-MIL-125 (Ti), Ti-MOF) has a high specific surface area of 1116 m2/g and excellent biocompatibility, and promotes osteogenic differentiation. The doxorubicin (DOX) loading capacity of Ti-MOF was 322 ± 21 mg/g, and DOX@Ti-MOF has perfect antitumor activity. Furthermore, the incorporation of DOX@Ti-MOF improved the physical and mechanical properties of the composite scaffolds, making the scaffold surface rough and favorable for cells to attach. CS/DOX@Ti-MOF retains the unique properties of each component. It responds to the release of DOX in the tumor microenvironment to remove residual tumor cells, followed by providing a site for cell attachment, proliferation, and differentiation. This promotes bone repair and achieves the sequential treatment of postoperative bone tumors. Overall, CS/DOX@Ti-MOF may be a promising substitute for postoperative bone tumor clearance and bone defect repair. It also provides a possible strategy for postoperative bone tumor treatment.


Asunto(s)
Neoplasias Óseas , Quitosano , Humanos , Osteogénesis , Titanio , Recurrencia Local de Neoplasia , Doxorrubicina/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/cirugía , Andamios del Tejido , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...