Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.882
Filtrar
1.
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
2.
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
3.
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
4.
J Nanobiotechnology ; 22(1): 577, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300539

RESUMEN

Treating bone defect concomitant with microbial infection poses a formidable clinical challenge. Addressing this dilemma necessitates the implementation of biomaterials exhibiting dual capabilities in anti-bacteria and bone regeneration. Of particular significance is the altered microenvironment observed in infected bones, characterized by acidity, inflammation, and an abundance of reactive oxygen species (ROS). These conditions, while challenging, present an opportunity for therapeutic intervention in the context of contaminated bone defects. In this study, we developed an oriented composite scaffold containing copper-coated manganese dioxide (MnO2) nanoparticles loaded with parathyroid hormone (PMPC/Gelatin). The characteristics of these scaffolds were meticulously evaluated and confirmed the high sensitivity to H+, responsive drug release and ROS elimination. In vitro antibacterial analysis underscored the remarkable ability of PMPC/Gelatin scaffolds to substantially suppressed bacterial proliferation and colony formation. Furthermore, this nontoxic material demonstrated efficacy in mitigating ROS levels, thereby fostering osteogenic differentiation of bone marrow mesenchymal stem cells and enhancing angiogenic ability. Subsequently, the infected models of bone defects in rat skulls were established to investigate the effects of composite scaffolds on anti-bacteria and bone formation in vivo. The PMPC/Gelatin treatment exhibited excellent antibacterial activity, coupled with enhanced vascularization and osteogenesis at the defect sites. These compelling findings affirm that the PMPC/Gelatin composite scaffold represents a promising avenue for anti-bacteria and bone regeneration.


Asunto(s)
Antibacterianos , Regeneración Ósea , Gelatina , Compuestos de Manganeso , Células Madre Mesenquimatosas , Osteogénesis , Óxidos , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Andamios del Tejido , Animales , Osteogénesis/efectos de los fármacos , Andamios del Tejido/química , Ratas , Regeneración Ósea/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/química , Células Madre Mesenquimatosas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Gelatina/química , Óxidos/química , Óxidos/farmacología , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Masculino , Cráneo/efectos de los fármacos , Nanopartículas/química , Cobre/química , Cobre/farmacología , Diferenciación Celular/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología
5.
Biomater Adv ; 165: 214010, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39222592

RESUMEN

The application of biomaterials in bone regeneration is a prevalent clinical practice. However, its efficacy in elderly patients remains suboptimal, necessitating further advancements. While biomaterial properties are known to orchestrate macrophage (MΦ) polarization and local immune responses, the role of biomaterial cues, specifically stiffness, in directing the senescent macrophage (S-MΦ) is still poorly understood. This study aimed to elucidate the role of substrate stiffness in modulating the immunomodulatory properties of S-MΦ and their role in osteo-immunomodulation. Our results demonstrated that employing collagen-coated polyacrylamide hydrogels with varying stiffness values (18, 76, and 295 kPa) as model materials, the high-stiffness hydrogel (295 kPa) steered S-MΦs towards a pro-inflammatory M1 phenotype, while hydrogels with lower stiffness (18 and 76 kPa) promoted an anti-inflammatory M2 phenotype. The immune microenvironment created by S-MΦs promoted the bioactivities of senescent endothelial cells (S-ECs) and senescent bone marrow mesenchymal stem cells BMSCs (S-BMSCs). Furthermore, the M2 S-MΦs, particularly incubated on the 76 kPa hydrogel matrices, significantly enhanced the ability of angiogenesis of S-ECs and osteogenic differentiation of S-BMSCs, which are crucial and interrelated processes in bone healing. This modulation aided in reducing the accumulation of reactive oxygen species in S-ECs and S-BMSCs, thereby significantly contributing to the repair and regeneration of aged bone tissue.


Asunto(s)
Regeneración Ósea , Hidrogeles , Inmunomodulación , Macrófagos , Células Madre Mesenquimatosas , Osteogénesis , Regeneración Ósea/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Hidrogeles/química , Osteogénesis/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Animales , Senescencia Celular/efectos de los fármacos , Humanos , Diferenciación Celular , Neovascularización Fisiológica/efectos de los fármacos , Resinas Acrílicas/química , Resinas Acrílicas/farmacología , Materiales Biocompatibles/farmacología , Propiedades de Superficie , Colágeno/metabolismo
6.
Int J Mol Sci ; 25(17)2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39273529

RESUMEN

Bone tissue regeneration is a critical aspect of dental surgery, given the common occurrence of bone resorption leading to alveolar bone defects. The aim of this paper was to conduct a systematic review to provide a comprehensive summary of the evidence regarding the regenerative properties of dentin biomaterial. This systematic review was conducted through comprehensive searches in the PubMed, Scopus, and Web of Science databases, as well as an extensive exploration of the gray literature sources, including WorldCat, The New York Academy of Medicine Library, and Trip Database, following the established PRISMA protocol. Keywords such as tooth, dentin, grinder, and autograft guided the search, with a focus on a standardized procedure involving dentin grinders within laboratory, experimental, and clinical settings. Initially, a pool of 1942 articles was identified with 452 duplicates removed. An additional 1474 articles were excluded for not aligning with the predefined topics, and three more were excluded due to the unavailability of the full text. Ultimately, 13 articles met the strict inclusion criteria and were included in the review. The chemical composition of the dentin particles was similar to natural bone in terms of oxygen, carbon, calcium, phosphorus, sodium, and magnesium content, as well as in terms of the Ca/P ratio. In addition, the dentin also contained amide I and amide II structures, as well as aliphatic and hydroxyl functional groups. The chemically treated dentin was free of microorganisms. The dentin had characteristic tubules that opened after chemical treatment. At the cellular level, dentin released bone morphogenetic protein 2, induced significant cell growth, and stimulated the reorganization of the fibroblast cytoskeleton. Most clinical studies have focused on alveolar bone regeneration. After the transplantation of demineralized dentin particles, studies have observed new bone formation, a reduction in residual bone, and an increase in connective tissue. Clinical reports consistently indicate uncomplicated healing and recovery post-transplantation. However, there is a notable gap in the evidence concerning complication rates, patient-reported outcomes, and the presence of pro-inflammatory factors. In conclusion, dentin biomaterial emerges as a versatile bone substitute, demonstrating high biocompatibility and ease of acquisition. The preservation of its internal structure containing organic matter and growth factors enhances its potential for effective bone regeneration. Particularly, in dental surgery, dentin-derived materials present a promising alternative to traditional autologous bone autografts, offering the potential to reduce patient morbidity and treatment costs.


Asunto(s)
Materiales Biocompatibles , Regeneración Ósea , Dentina , Dentina/metabolismo , Humanos , Regeneración Ósea/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Animales
7.
BMC Vet Res ; 20(1): 403, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251976

RESUMEN

BACKGROUND: The integration of stem cells, signaling molecules, and biomaterial scaffolds is fundamental for the successful engineering of functional bone tissue. Currently, the development of composite scaffolds has emerged as an attractive approach to meet the criteria of ideal scaffolds utilized in bone tissue engineering (BTE) for facilitating bone regeneration in bone defects. Recently, the incorporation of polycaprolactone (PCL) with hydroxyapatite (HA) has been developed as one of the suitable substitutes for BTE applications owing to their promising osteogenic properties. In this study, a three-dimensional (3D) scaffold composed of PCL integrated with HA (PCL/HA) was prepared and assessed for its ability to support osteogenesis in vitro. Furthermore, this scaffold was evaluated explicitly for its efficacy in promoting the proliferation and osteogenic differentiation of canine bone marrow-derived mesenchymal stem cells (cBM-MSCs) to fill the knowledge gap regarding the use of composite scaffolds for BTE in the veterinary orthopedics field. RESULTS: Our findings indicate that the PCL/HA scaffolds substantially supported the proliferation of cBM-MSCs. Notably, the group subjected to osteogenic induction exhibited a markedly upregulated expression of the osteogenic gene osterix (OSX) compared to the control group. Additionally, the construction of 3D scaffold constructs with differentiated cells and an extracellular matrix (ECM) was successfully imaged using scanning electron microscopy. Elemental analysis using a scanning electron microscope coupled with energy-dispersive X-ray spectroscopy confirmed that these constructs possessed the mineral content of bone-like compositions, particularly the presence of calcium and phosphorus. CONCLUSIONS: This research highlights the synergistic potential of PCL/HA scaffolds in concert with cBM-MSCs, presenting a multidisciplinary approach to scaffold fabrication that effectively regulates cell proliferation and osteogenic differentiation. Future in vivo studies focusing on the repair and regeneration of bone defects are warranted to further explore the regenerative capacity of these constructs, with the ultimate goal of assessing their potential in veterinary clinical applications.


Asunto(s)
Regeneración Ósea , Durapatita , Células Madre Mesenquimatosas , Osteogénesis , Poliésteres , Andamios del Tejido , Animales , Perros , Poliésteres/química , Poliésteres/farmacología , Andamios del Tejido/química , Osteogénesis/efectos de los fármacos , Durapatita/química , Durapatita/farmacología , Células Madre Mesenquimatosas/fisiología , Regeneración Ósea/efectos de los fármacos , Proliferación Celular , Diferenciación Celular/efectos de los fármacos , Ingeniería de Tejidos/métodos
8.
J Nanobiotechnology ; 22(1): 539, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39237993

RESUMEN

Maxillofacial bone defects can severely impact quality of life by impairing physiological functions such as chewing, breathing, swallowing, and pronunciation. Polyether ether ketone (PEEK) is commonly used for the repair of maxillofacial defects due to its mechanical adaptability, while its osteogenic properties still need refinement. Herein, we have utilized the piezoelectric effect exhibited by barium titanate (BTO) under low-intensity pulsed ultrasound (LIPUS) to develop an ultrasound responsive PEEK (PDA@BTO-SPEEK, PBSP) through the mediating effect of polydopamine (PDA), for repairing maxillofacial bone defects. After modification by PDA@BTO, PBSP possesses better hydrophilicity, which is conducive to cell growth and adhesion. Simultaneously, by virtue of the piezoelectric characteristics of BTO, PBSP obtains a piezoelectric coefficient that matches the bone cortex. Notably, when PBSP is stimulated by LIPUS, it can generate stable electricity and effectively accelerate the osteogenic differentiation of osteoblasts through the regulation of the Piezo1-induced calcium (Ca2+) influx and Akt/GSK3ß/ß-catenin pathway. In addition, PBSP presents satisfactory therapeutic effects in rat skull defect models, and its osteogenic efficiency can be further improved under LIPUS stimulation with high tissue penetration. Collectively, PBSP + LIPUS exhibits great potential as a promising alternative strategy for the repair of maxillofacial bone defects.


Asunto(s)
Benzofenonas , Glucógeno Sintasa Quinasa 3 beta , Cetonas , Osteogénesis , Polietilenglicoles , Polímeros , Proteínas Proto-Oncogénicas c-akt , Ratas Sprague-Dawley , beta Catenina , Animales , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Polímeros/química , Osteogénesis/efectos de los fármacos , Ratas , Polietilenglicoles/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Cetonas/química , Cetonas/farmacología , beta Catenina/metabolismo , Diferenciación Celular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Ondas Ultrasónicas , Indoles/química , Indoles/farmacología , Masculino , Transducción de Señal/efectos de los fármacos , Cráneo/efectos de los fármacos , Titanio/química , Titanio/farmacología , Regeneración Ósea/efectos de los fármacos
9.
ACS Appl Mater Interfaces ; 16(36): 47178-47191, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39222394

RESUMEN

Guided bone regeneration (GBR) technology has been demonstrated to be an effective method for reconstructing bone defects. A membrane is used to cover the bone defect to stop soft tissue from growing into it. The biosurface design of the barrier membrane is key to the technology. In this work, an asymmetric functional gradient Janus membrane was designed to address the bidirectional environment of the bone and soft tissue during bone reconstruction. The Janus membrane was simply and efficiently prepared by the multilayer self-assembly technique, and it was divided into the polycaprolactone isolation layer (PCL layer, GBR-A) and the nanohydroxyapatite/polycaprolactone/polyethylene glycol osteogenic layer (HAn/PCL/PEG layer, GBR-B). The morphology, composition, roughness, hydrophilicity, biocompatibility, cell attachment, and osteogenic mineralization ability of the double surfaces of the Janus membrane were systematically evaluated. The GBR-A layer was smooth, dense, and hydrophobic, which could inhibit cell adhesion and resist soft tissue invasion. The GBR-B layer was rough, porous, hydrophilic, and bioactive, promoting cell adhesion, proliferation, matrix mineralization, and expression of alkaline phosphatase and RUNX2. In vitro and in vivo results showed that the membrane could bind tightly to bone, maintain long-term space stability, and significantly promote new bone formation. Moreover, the membrane could fix the bone filling material in the defect for a better healing effect. This work presents a straightforward and viable methodology for the fabrication of GBR membranes with Janus-based bioactive surfaces. This work may provide insights for the design of biomaterial surfaces and treatment of bone defects.


Asunto(s)
Regeneración Ósea , Osteogénesis , Poliésteres , Regeneración Ósea/efectos de los fármacos , Animales , Poliésteres/química , Poliésteres/farmacología , Osteogénesis/efectos de los fármacos , Durapatita/química , Durapatita/farmacología , Polietilenglicoles/química , Membranas Artificiales , Adhesión Celular/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Proliferación Celular/efectos de los fármacos , Humanos , Regeneración Tisular Dirigida/métodos , Conejos , Ratones
10.
ACS Appl Mater Interfaces ; 16(37): 49135-49147, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39226455

RESUMEN

The treatment of irregular-shaped and critical-sized bone defects poses a clinical challenge. Deployable, self-fitting tissue scaffolds that can be implanted by minimally invasive procedures are a promising solution. Toward this, we fabricated NIR-responsive and programmable polylactide-co-trimethylene carbonate (PLMC) scaffolds nanoengineered with polydopamine nanoparticles (PDA) by extrusion-based three-dimensional (3D) printing. The 3D-printed scaffolds demonstrated excellent (>99%), fast (under 30 s), and tunable shape recovery under NIR irradiation. PLMC-PDA composites demonstrated significantly higher osteogenic potential in vitro as revealed by the significantly enhanced alkaline phosphatase (ALP) secretion and mineral deposition in contrast to neat PLMC. Intraoperative deployability and in vivo bone regeneration ability of PLMC-PDA composites were demonstrated, using self-fitting scaffolds in critical-sized cranial bone defects in rabbits. The 3D-printed scaffolds were deformed into compact shapes that could self-fit into the defect shape intraoperatively under low power intensity (0.76 W cm-2) NIR. At 6 and 12 weeks postsurgical implantation, near-complete bone regeneration was observed in PLMC-PDA composites, unlike neat PLMC through microcomputed tomography (micro-CT) analysis. The potential clinical utility of the 3D-printed composites to secure complex defects was confirmed through self-fitting of the scaffolds into irregular defects in ex vivo models of rabbit tibia, mandible, and tooth models. Taken together, the composite scaffolds fabricated here offer an innovative strategy for minimally invasive deployment to fit irregular and complex tissue defects for bone tissue regeneration.


Asunto(s)
Regeneración Ósea , Indoles , Osteogénesis , Polímeros , Impresión Tridimensional , Andamios del Tejido , Animales , Andamios del Tejido/química , Conejos , Regeneración Ósea/efectos de los fármacos , Polímeros/química , Indoles/química , Osteogénesis/efectos de los fármacos , Poliésteres/química , Nanopartículas/química , Ingeniería de Tejidos , Dioxanos/química , Rayos Infrarrojos , Huesos/diagnóstico por imagen , Huesos/patología
11.
J Biomed Mater Res B Appl Biomater ; 112(10): e35486, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39295151

RESUMEN

Hydrogels have emerged as potential materials for bone grafting, thanks to their biocompatibility, biodegradation, and flexibility in filling irregular bone defects. In this study, we fabricated a novel NAH hydrogel system, composed of N,O-carboxymethyl chitosan (NOCC), aldehyde hyaluronic acid (AHA), and hydroxyapatite (HAp). To improve the mechanical strength of the fabricated hydrogel, a porous polycaprolactone (PCL) matrix was synthesized and used as a three-dimensional (3D) support template for NAH hydrogel loading, forming a novel PCL/NAH hybrid scaffold. A mixture of monosodium glutamate (M) and sucrose (S) at varied weight ratios (5M:5S, 7M:3S, and 9M:1S) was used for the fabrication of 3D PCL matrices. The morphology, interconnectivity, and water resistance of the porous PCL scaffolds were investigated for optimal hydrogel loading efficiency. The results demonstrated that PCL scaffolds with porogen ratios of 7M:3S and 9M:1S possessed better interconnectivity than 5M:5S ratio. The compressive strength of the PCL/NAH hybrid scaffolds with 9M:1S (561.6 ± 6.1 kPa) and 7M:3S (623.8 ± 6.8 kPa) ratios are similar to cancellous bone and all hybrid scaffolds were biocompatible. Rabbit models with tibial defects were implanted with the PCL/NAH scaffolds to assess the wound healing capability. The results suggest that the PCL/NAH hybrid scaffolds, specifically those with porogen ratio of 7M:3S, exhibit promising bone healing effects.


Asunto(s)
Regeneración Ósea , Quitosano , Durapatita , Ácido Hialurónico , Hidrogeles , Poliésteres , Andamios del Tejido , Quitosano/química , Quitosano/farmacología , Quitosano/análogos & derivados , Animales , Conejos , Durapatita/química , Durapatita/farmacología , Andamios del Tejido/química , Regeneración Ósea/efectos de los fármacos , Poliésteres/química , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Ensayo de Materiales , Masculino
12.
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
13.
Int J Oral Implantol (Berl) ; 17(3): 237-248, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39283217

RESUMEN

Dental implants are a reliable treatment option for restoring missing teeth, but adequate bone quantity and quality are crucial for success. This case series presents four cases treated by different clinicians, all following very similar concepts for combined periodontal and vertical ridge augmentation using recombinant human platelet-derived growth factor-BB. All cases involved a severe periodontal defect requiring either extraction of the adjacent tooth or periodontal regeneration. Different bone grafts and membrane types were utilised. Although true periodontal regeneration cannot be said categorically to have occurred due to a lack of histological evidence, the clinical and radiographic findings suggest almost complete bone fill in all cases. This case series demonstrates that combined periodontal and vertical ridge augmentation using recombinant human platelet-derived growth factor-BB could be successful, but proper case selection and patient preparation for the possibility of multiple surgical procedures are recommended. Conflict-of-interest statement: At the time of preparing this manuscript, Dr Saleh was a clinical advisor for Lynch Biologics, Franklin, TN, USA. The other authors declare that they have no conflicts of interest relating to this study.


Asunto(s)
Aumento de la Cresta Alveolar , Becaplermina , Humanos , Femenino , Masculino , Aumento de la Cresta Alveolar/métodos , Persona de Mediana Edad , Becaplermina/uso terapéutico , Adulto , Proteínas Recombinantes/uso terapéutico , Regeneración Ósea/efectos de los fármacos , Pérdida de Hueso Alveolar/cirugía , Pérdida de Hueso Alveolar/tratamiento farmacológico , Trasplante Óseo/métodos , Proteínas Proto-Oncogénicas c-sis/uso terapéutico , Regeneración Tisular Guiada Periodontal/métodos
14.
Molecules ; 29(17)2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39275056

RESUMEN

Bone has the ability to heal itself; however, bone defects fail to heal once the damage exceeds a critical size. Bone regeneration remains a significant clinical challenge, with autograft considered the ideal bone graft material due to its sufficient porosity, osteogenic cells, and biological growth factors. However, limitations to bone grafting, such as limited bone stock and high resorption rates, have led to a great deal of research into developing bone graft substitutes. The P28 peptide is a small molecule bioactive biomimetic alternative to mimic the bone morphogenetic protein 2 (BMP-2). In this study, we investigated the potential of P28-loaded hybrid scaffolds to mimic the natural bone structure for enhancing the bone regeneration process. We hypothesized that the peptide-loaded scaffolds and nude scaffolds both have the potential to promote bone healing, and the bone healing process is accelerated by the release of the peptide. To verify our hypothesis, C2C12 cells were evaluated for the presence of calcium deposits by histological stain at 7 and 14 days in cultures with hybrid scaffolds. Total RNA was isolated from C2C12 cells cultured with hybrid scaffolds for 7 and 14 days to assess osteoblast differentiation. The project findings demonstrated that the hybrid scaffold could enhance osteoblast differentiation and significantly improve the therapeutic effects of the scaffold in bone regeneration.


Asunto(s)
Regeneración Ósea , Diferenciación Celular , Cerámica , Quitosano , Andamios del Tejido , Regeneración Ósea/efectos de los fármacos , Andamios del Tejido/química , Quitosano/química , Quitosano/farmacología , Animales , Ratones , Cerámica/química , Cerámica/farmacología , Diferenciación Celular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/metabolismo , Línea Celular , Osteogénesis/efectos de los fármacos , Proteína Morfogenética Ósea 2/farmacología , Ingeniería de Tejidos/métodos , Péptidos/química , Péptidos/farmacología , Humanos
15.
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
16.
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
17.
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
18.
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
19.
Biomacromolecules ; 25(9): 5512-5540, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39133748

RESUMEN

Current bone repair methods have limitations, prompting the exploration of innovative approaches. Tissue engineering emerges as a promising solution, leveraging biomaterials to craft scaffolds replicating the natural bone environment, facilitating cell growth and differentiation. Among fabrication techniques, three-dimensional (3D) printing stands out for its ability to tailor intricate scaffolds. Silk proteins (SPs), known for their mechanical strength and biocompatibility, are an excellent choice for engineering 3D-printed bone tissue engineering (BTE) scaffolds. This article comprehensively reviews bone biology, 3D printing, and the unique attributes of SPs, specifically detailing criteria for scaffold fabrication such as composition, structure, mechanics, and cellular responses. It examines the structural, mechanical, and biological attributes of SPs, emphasizing their suitability for BTE. Recent studies on diverse 3D printing approaches using SPs-based for BTE are highlighted, alongside advancements in their 3D and four-dimensional (4D) printing and their role in osteo-immunomodulation. Future directions in the use of SPs for 3D printing in BTE are outlined.


Asunto(s)
Regeneración Ósea , Inmunomodulación , Impresión Tridimensional , Seda , Ingeniería de Tejidos , Andamios del Tejido , Regeneración Ósea/efectos de los fármacos , Humanos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Seda/química , Animales , Inmunomodulación/efectos de los fármacos , Materiales Biocompatibles/química , Huesos/metabolismo
20.
ACS Biomater Sci Eng ; 10(9): 5723-5738, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39163588

RESUMEN

The repair and regeneration of maxillofacial bone defects are major clinical challenges. Titanium (Ti)-magnesium (Mg) composites are a new generation of revolutionary internal fixation materials encompassing the mechanical strength and bioactive advantages of Ti and Mg alloys, respectively. This study was aimed to construct a Ti-Mg composite internal plate/screw fixation system to fix and repair bone defects. Further, the effects of different internal fixation systems on bone repair were analyzed through radiological and histological analyses. Notably, Ti6Al4V with rolled Mg foil was used as the experimental group, and a bone defect model of transverse complete amputation of the ulna in rabbits similar to the clinical condition was established. The internal fixation system with the highest osteogenic efficiency was selected based on in vivo results, and the direct and indirect bone repair abilities of the selected materials were evaluated in vitro. Notably, the thin Mg foil-Ti6Al4V internal fixation system exhibited the best fixation effect in the bone defect model and promoted the formation of new bone and early healing of bone defect areas. In vitro, the thin Mg foil-Ti6Al4V composite enhanced the activity of MC3T3-E1 cells; promoted the proliferation, adhesion, extension, and osteogenic differentiation of MC3T3-E1 cells; and regulated new bone formation. Further, it also promoted the polarization of RAW264.7 cells to M2 macrophages, induced the osteogenic immune microenvironment, and indirectly regulated the bone repair process. Therefore, a internal fixation system holds a promising potential for the internal fixation of maxillofacial bone defects. Our findings provide a theoretical and scientific basis for the design and clinical application of Ti-Mg internal fixation systems.


Asunto(s)
Aleaciones , Magnesio , Osteogénesis , Titanio , Animales , Titanio/química , Magnesio/farmacología , Conejos , Ratones , Aleaciones/química , Osteogénesis/efectos de los fármacos , Regeneración Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Fijación Interna de Fracturas/métodos , Diferenciación Celular/efectos de los fármacos , Fijadores Internos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA