Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
ACS Appl Mater Interfaces ; 16(37): 49135-49147, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39226455

RESUMO

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.


Assuntos
Regeneração Óssea , Indóis , Osteogênese , Polímeros , Impressão Tridimensional , Alicerces Teciduais , Animais , Alicerces Teciduais/química , Coelhos , Regeneração Óssea/efeitos dos fármacos , Polímeros/química , Indóis/química , Osteogênese/efeitos dos fármacos , Poliésteres/química , Nanopartículas/química , Engenharia Tecidual , Dioxanos/química , Raios Infravermelhos , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia
2.
Drug Deliv ; 31(1): 2391001, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39239763

RESUMO

A common malignant bone neoplasm in teenagers is Osteosarcoma. Chemotherapy, surgical therapy, and radiation therapy together comprise the usual clinical course of treatment for Osteosarcoma. While Osteosarcoma and other bone tumors are typically treated surgically, however, surgical resection frequently fails to completely eradicate tumors, and in turn becomes the primary reason for postoperative recurrence and metastasis, ultimately leading to a high rate of mortality. Patients still require radiation and/or chemotherapy after surgery to stop the spread of the tumor and its metastases, and both treatments have an adverse influence on the body's organ systems. In the postoperative management of osteosarcoma, bone scaffolds can load cargos (growth factors or drugs) and function as drug delivery systems (DDSs). This review describes the different kinds of bone scaffolds that are currently available and highlights key studies that use scaffolds as DDSs for the treatment of osteosarcomas. The discussion also includes difficulties and perspectives regarding the use of scaffold-based DDSs. The study may serve as a source for outlining efficient and secure postoperative osteosarcoma treatment plans.


Assuntos
Neoplasias Ósseas , Sistemas de Liberação de Medicamentos , Osteossarcoma , Alicerces Teciduais , Osteossarcoma/tratamento farmacológico , Humanos , Sistemas de Liberação de Medicamentos/métodos , Neoplasias Ósseas/tratamento farmacológico , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Osso e Ossos/efeitos dos fármacos , Animais
3.
J Nanobiotechnology ; 22(1): 500, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39169401

RESUMO

Bone defects pose significant challenges in healthcare, with over 2 million bone repair surgeries performed globally each year. As a burgeoning force in the field of bone tissue engineering, 3D printing offers novel solutions to traditional bone transplantation procedures. However, current 3D-printed bone scaffolds still face three critical challenges in material selection, printing methods, cellular self-organization and co-culture, significantly impeding their clinical application. In this comprehensive review, we delve into the performance criteria that ideal bone scaffolds should possess, with a particular focus on the three core challenges faced by 3D printing technology during clinical translation. We summarize the latest advancements in non-traditional materials and advanced printing techniques, emphasizing the importance of integrating organ-like technologies with bioprinting. This combined approach enables more precise simulation of natural tissue structure and function. Our aim in writing this review is to propose effective strategies to address these challenges and promote the clinical translation of 3D-printed scaffolds for bone defect treatment.


Assuntos
Bioimpressão , Osso e Ossos , Organoides , Impressão Tridimensional , Engenharia Tecidual , Alicerces Teciduais , Alicerces Teciduais/química , Humanos , Engenharia Tecidual/métodos , Organoides/citologia , Bioimpressão/métodos , Animais , Regeneração Óssea , Transplante Ósseo/métodos
4.
Int J Nanomedicine ; 19: 8309-8336, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39161358

RESUMO

Purpose: The treatment of craniofacial bone defects caused by trauma, tumors, and infectious and degenerative diseases is a significant issue in current clinical practice. Following the rapid development of bone tissue engineering (BTE) in the last decade, bioactive scaffolds coupled with multifunctional properties are in high demand with regard to effective therapy for bone defects. Herein, an innovative bone scaffold consisting of GO/Cu nanoderivatives and GelMA-based organic-inorganic hybrids was reported for repairing full-thickness calvarial bone defect. Methods: In this study, motivated by the versatile biological functions of nanomaterials and synthetic hydrogels, copper nanoparticle (CuNP)-decorated graphene oxide (GO) nanosheets (GO/Cu) were combined with methacrylated gelatin (GelMA)-based organic-inorganic hybrids to construct porous bone scaffolds that mimic the extracellular matrix (ECM) of bone tissues by photocrosslinking. The material characterizations, in vitro cytocompatibility, macrophage polarization and osteogenesis of the biohybrid hydrogel scaffolds were investigated, and two different animal models (BALB/c mice and SD rats) were established to further confirm the in vivo neovascularization, macrophage recruitment, biocompatibility, biosafety and bone regenerative potential. Results: We found that GO/Cu-functionalized GelMA/ß-TCP hydrogel scaffolds exhibited evidently promoted osteogenic activities, M2 type macrophage polarization, increased secretion of anti-inflammatory factors and excellent cytocompatibility, with favorable surface characteristics and sustainable release of Cu2+. Additionally, improved neovascularization, macrophage recruitment and tissue integration were found in mice implanted with the bioactive hydrogels. More importantly, the observations of microCT reconstruction and histological analysis in a calvarial bone defect model in rats treated with GO/Cu-incorporated hydrogel scaffolds demonstrated significantly increased bone morphometric values and newly formed bone tissues, indicating accelerated bone healing. Conclusion: Taken together, this BTE-based bone repair strategy provides a promising and feasible method for constructing multifunctional GO/Cu nanocomposite-incorporated biohybrid hydrogel scaffolds with facilitated osteogenesis, angiogenesis and immunoregulation in one system, with the optimization of material properties and biosafety, it thereby demonstrates great application potential for correcting craniofacial bone defects in future clinical scenarios.


Assuntos
Regeneração Óssea , Cobre , Grafite , Hidrogéis , Ratos Sprague-Dawley , Crânio , Engenharia Tecidual , Alicerces Teciduais , Animais , Regeneração Óssea/efeitos dos fármacos , Alicerces Teciduais/química , Cobre/química , Cobre/farmacologia , Grafite/química , Hidrogéis/química , Hidrogéis/farmacologia , Crânio/efeitos dos fármacos , Crânio/lesões , Ratos , Camundongos , Engenharia Tecidual/métodos , Osteogênese/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Masculino , Nanopartículas Metálicas/química , Nanoestruturas/química , Gelatina/química , Células RAW 264.7
5.
Int J Biol Macromol ; 279(Pt 2): 135182, 2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39216566

RESUMO

We have fabricated and characterized novel bioactive nanocomposite interpenetrating polymer network (IPN) scaffolds to treat bone defects by loading mesoporous silica nanoparticles (MSNs) into blends of Konjac glucomannan, polyvinyl alcohol, and polycaprolactone. By loading MSNs, we developed a porous nanocomposite scaffold with mechanical strengths comparable to cancellous bone. In vitro cell culture studies proved the cytocompatibility of the nanocomposite scaffolds. RT-PCR studies confirmed that these scaffolds significantly upregulated major osteogenic markers. The in vivo chick chorioallantoic membrane (CAM) assay confirmed the proangiogenic activity of the nanocomposite IPN scaffolds. In vivo studies were performed using Wistar rats to evaluate the scaffolds' compatibility, osteogenic activity, and proangiogenic properties. Liver and renal function tests confirmed that these scaffolds were nontoxic. X-ray and µ-CT results show that the bone defects treated with the nanocomposite scaffolds healed at a much faster rate compared to the untreated control and those treated with IPN scaffolds. H&E and Masson's trichrome staining showed angiogenesis near the newly formed bone and the presence of early-stage connective tissues, fibroblasts, and osteoblasts in the defect region at 8 weeks after surgery. Hence, these advantageous physicochemical and biological properties confirm that the nanocomposite IPN scaffolds are ideal for treating bone defects.

6.
Adv Sci (Weinh) ; 11(35): e2401589, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39018263

RESUMO

Using bone regeneration scaffolds to repair craniomaxillofacial bone defects is a promising strategy. However, most bone regeneration scaffolds still exist some issues such as a lack of barrier structure, inability to precisely match bone defects, and necessity to incorporate biological components to enhance efficacy. Herein, inspired by a periosteum-bone complex, a class of multifunctional hierarchical porous poly(lactic-co-glycolic acid)/baicalein scaffolds is facilely prepared by the union of personalized negative mold technique and phase separation strategy and demonstrated to precisely fit intricate bone defect cavity. The dense up-surface of the scaffold can prevent soft tissue cell penetration, while the loose bottom-surface can promote protein adsorption, cell adhesion, and cell infiltration. The interior macropores of the scaffold and the loaded baicalein can synergistically promote cell differentiation, angiogenesis, and osteogenesis. These findings can open an appealing avenue for the development of personalized multifunctional hierarchical materials for bone repair.


Assuntos
Regeneração Óssea , Periósteo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Engenharia Tecidual , Alicerces Teciduais , Alicerces Teciduais/química , Regeneração Óssea/fisiologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Animais , Porosidade , Engenharia Tecidual/métodos , Osteogênese/fisiologia , Diferenciação Celular , Humanos , Modelos Animais de Doenças , Materiais Biocompatíveis/química , Camundongos
7.
J Biomed Mater Res A ; 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38864151

RESUMO

Infection is a major concern in surgery involving grafting and should be considered thoroughly when designing biomaterials. There is considerable renewed interest in silver nanoparticles (AgNPs) owing to their ability to potentiate antibacterial properties against multiple bacterial strains. This study aimed to develop two antibacterial bone regenerative scaffolds by integrating AgNPs in bovine bone particles (BBX) (Product 1), and a light cross-linked hydrogel GelMA (Product 2). The constructs were characterized using scanning electron microscopy. Metabolic activity of osteoblasts and osteoclasts on the constructs was investigated using PrestoBlue™. Disk diffusion assay was conducted to test the antibacterial properties. The regenerative capacity of the optimized AgNP functionalized BBX and GelMA were tested in a rabbit cranial 6 mm defect model. The presence of AgNPs appears to enhance proliferation of osteoblasts compared to AgNP free controls in vitro. We established that AgNPs can be used at a 100 µg dose that inhibits bacteria, with minimal adverse effects on the bone cells. Our rabbit model revealed that both the BBX and GelMA hydrogels loaded AgNPs were biocompatible with no signs of necrosis or inflammatory response. Grafts functionalized with AgNPs can provide antibacterial protection and simultaneously act as a scaffold for attachment of bone cells.

8.
ACS Appl Bio Mater ; 7(7): 4366-4378, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38905196

RESUMO

Bone is remodeled through a dynamic process facilitated by biophysical cues that support cellular signaling. In healthy bone, signaling pathways are regulated by cells and the extracellular matrix and transmitted via electrical synapses. To this end, combining electrical stimulation (ES) with conductive scaffolding is a promising approach for repairing damaged bone tissue. Therefore, "smart" biomaterials that can provide multifunctionality and facilitate the transfer of electrical cues directly to cells have become increasingly more studied in bone tissue engineering. Herein, 3D-printed electrically conductive composite scaffolds consisting of demineralized bone matrix (DBM) and polycaprolactone (PCL), in combination with ES, for bone regeneration were evaluated for the first time. The conductive composite scaffolds were fabricated and characterized by evaluating mechanical, surface, and electrical properties. The DBM/PCL composites exhibited a higher compressive modulus (107.2 MPa) than that of pristine PCL (62.02 MPa), as well as improved surface properties (i.e., roughness). Scaffold electrical properties were also tuned, with sheet resistance values as low as 4.77 × 105 Ω/sq for our experimental coating of the highest dilution (i.e., 20%). Furthermore, the biocompatibility and osteogenic potential of the conductive composite scaffolds were tested using human mesenchymal stromal cells (hMSCs) both with and without exogenous ES (100 mV/mm for 5 min/day four times/week). In conjunction with ES, the osteogenic differentiation of hMSCs grown on conductive DBM/PCL composite scaffolds was significantly enhanced when compared to those cultured on PCL-only and nonconductive DBM/PCL control scaffolds, as determined through xylenol orange mineral staining and osteogenic protein analysis. Overall, these promising results suggest the potential of this approach for the development of biomimetic hybrid scaffolds for bone tissue engineering applications.


Assuntos
Materiais Biocompatíveis , Matriz Óssea , Estimulação Elétrica , Teste de Materiais , Impressão Tridimensional , Engenharia Tecidual , Alicerces Teciduais , Alicerces Teciduais/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Humanos , Matriz Óssea/química , Condutividade Elétrica , Poliésteres/química , Osteogênese , Tamanho da Partícula , Células-Tronco Mesenquimais/citologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-38943424

RESUMO

The effective reconstruction of osteochondral biomimetic structures is a key factor in guiding the regeneration of full-thickness osteochondral defects. Due to the avascular nature of hyaline cartilage, the greatest challenge in constructing this scaffold lies in both utilizing the biomimetic structure to promote vascular differentiation for nutrient delivery to hyaline cartilage, thereby enhancing the efficiency of osteochondral reconstruction, and effectively blocking vascular ingrowth into the cartilage layer to prevent cartilage mineralization. However, the intrinsic relationship between the planning of the microporous pipe network and the flow resistance in the biomimetic structure, and the mechanism of promoting cell adhesion to achieve vascular differentiation and inhibiting cell adhesion to block the growth of blood vessels are still unclear. Inspired by the structure of tree trunks, this study designed a biomimetic tree-like tubular network structure for osteochondral scaffolds based on Murray's law. Utilizing computational fluid dynamics, the study investigated the influence of the branching angle of micro-pores on the flow velocity, pressure distribution, and scaffold permeability within the scaffold. The results indicate that when the differentiation angle exceeds 50 degrees, the highest flow velocity occurs at the confluence of tributaries at the ninth fractal position, forming a barrier layer. This structure effectively guides vascular growth, enhances nutrient transport capacity, increases flow velocity to promote cell adhesion, and inhibits cell infiltration into the cartilage layer.

10.
Bioprocess Biosyst Eng ; 47(9): 1483-1498, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38869621

RESUMO

Biosilica (BS) and spongin (SPG) from marine sponges are highlighted for their potential to promote bone regeneration. Moreover, 3D printing is introduced as a technology for producing bone grafts with optimized porous structures, allowing for better cell attachment, proliferation, and differentiation. Thus, this study aimed to characterize the BS and BS/SPG 3D printed scaffolds and to evaluate the biological effects in vitro. The scaffolds were printed using an ink containing 4 wt.% of sodium alginate. The physicochemical characteristics of BS and BS/SPG 3D printed scaffolds were analyzed by SEM, EDS, FTIR, porosity, evaluation of mass loss, and pH measurement. For in vitro analysis, the cellular viability of the MC3T3-E1 cell lineage was assessed using the AlamarBlue® assay and confocal microscopy, while genotoxicity and mineralization potential were evaluated through the micronucleus assay and Alizarin Red S, respectively. SEM analysis revealed spicules in BS, the fibrillar structure of SPG, and material degradation over the immersion period. FTIR indicated peaks corresponding to silicon oxide in BS samples and carbon oxide and amine in SPG samples. BS-SPG scaffolds exhibited higher porosity, while BS scaffolds displayed greater mass loss. pH measurements indicated a significant decrease induced by BS, which was mitigated by SPG over the experimental periods. In vitro studies demonstrated the biocompatibility and non-cytotoxicity of scaffold extracts. .Also, the scaffolds promoted cellular differentiation. The micronucleus test further confirmed the absence of genotoxicity. These findings suggest that 3D printed BS and BS/SPG scaffolds may possess desirable morphological and physicochemical properties, indicating in vitro biocompatibility.


Assuntos
Poríferos , Impressão Tridimensional , Alicerces Teciduais , Animais , Alicerces Teciduais/química , Poríferos/química , Camundongos , Dióxido de Silício/química , Regeneração Óssea , Porosidade , Sobrevivência Celular , Engenharia Tecidual/métodos , Linhagem Celular , Osso e Ossos
11.
Front Bioeng Biotechnol ; 12: 1372636, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38707506

RESUMO

Repair of large bone defects remains challenge for orthopedic clinical treatment. Porous titanium alloys have been widely fabricated by the additive manufacturing, which possess the elastic modulus close to that of human cortical bone, good osteoconductivity and osteointegration. However, insufficient bone regeneration and vascularization inside the porous titanium scaffolds severely limit their capability for repair of large-size bone defects. Therefore, it is crucially important to improve the osteogenic function and vascularization of the titanium scaffolds. Herein, methacrylated gelatin (GelMA) were incorporated with the porous Ti-24Nb-4Zr-8Sn (Ti2448) scaffolds prepared by the electron beam melting (EBM) method (Ti2448-GelMA). Besides, the deferoxamine (DFO) as an angiogenic agent was doped into the Ti2448-GelMA scaffold (Ti2448-GelMA/DFO), in order to promote vascularization. The results indicate that GelMA can fully infiltrate into the pores of Ti2448 scaffolds with porous cross-linked network (average pore size: 120.2 ± 25.1 µm). Ti2448-GelMA scaffolds facilitated the differentiation of MC3T3-E1 cells by promoting the ALP expression and mineralization, with the amount of calcium contents ∼2.5 times at day 14, compared with the Ti2448 scaffolds. Impressively, the number of vascular meshes for the Ti2448-GelMA/DFO group (∼7.2/mm2) was significantly higher than the control group (∼5.3/mm2) after cultivation for 9 h, demonstrating the excellent angiogenesis ability. The Ti2448-GelMA/DFO scaffolds also exhibited sustained release of DFO, with a cumulative release of 82.3% after 28 days. Therefore, Ti2448-GelMA/DFO scaffolds likely provide a new strategy to improve the osteogenesis and angiogenesis for repair of large bone defects.

12.
Acta Biomater ; 180: 115-127, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38642786

RESUMO

Bone has the capacity to regenerate itself for relatively small defects; however, this regenerative capacity is diminished in critical-size bone defects. The development of synthetic materials has risen as a distinct strategy to address this challenge. Effective synthetic materials to have emerged in recent years are bioceramic implants, which are biocompatible and highly bioactive. Yet nothing suitable for the repair of large bone defects has made the transition from laboratory to clinic. The clinical success of bioceramics has been shown to depend not only on the scaffold's intrinsic material properties but also on its internal porous geometry. This study aimed to systematically explore the implications of varying channel size, shape, and curvature in tissue scaffolds on in vivo bone regeneration outcomes. 3D printed bioceramic scaffolds with varying channel sizes (0.3 mm to 1.5 mm), shapes (circular vs rectangular), and curvatures (concave vs convex) were implanted in rabbit femoral defects for 8 weeks, followed by histological evaluation. We demonstrated that circular channel sizes of around 0.9 mm diameter significantly enhanced bone formation, compared to channel with diameters of 0.3 mm and 1.5 mm. Interestingly, varying channel shapes (rectangular vs circular) had no significant effect on the volume of newly formed bone. Furthermore, the present study systematically demonstrated the beneficial effect of concave surfaces on bone tissue growth in vivo, reinforcing previous in silico and in vitro findings. This study demonstrates that optimizing architectural configurations within ceramic scaffolds is crucial in enhancing bone regeneration outcomes. STATEMENT OF SIGNIFICANCE: Despite the explosion of work on developing synthetic scaffolds to repair bone defects, the amount of new bone formed by scaffolds in vivo remains suboptimal. Recent studies have illuminated the pivotal role of scaffolds' internal architecture in osteogenesis. However, these investigations have mostly remained confined to in silico and in vitro experiments. Among the in vivo studies conducted, there has been a lack of systematic analysis of individual architectural features. Herein, we utilized bioceramic 3D printing to conduct a systematic exploration of the effects of channel size, shape, and curvature on bone formation in vivo. Our results demonstrate the significant influence of channel size and curvature on in vivo outcomes. These findings provide invaluable insights into the design of more effective bone scaffolds.


Assuntos
Cerâmica , Osteogênese , Alicerces Teciduais , Impressão Tridimensional , Cerâmica/química , Alicerces Teciduais/química , Alicerces Teciduais/normas , Osteogênese/fisiologia , Animais , Coelhos , Masculino , Propriedades de Superfície
13.
Ann Biomed Eng ; 52(6): 1518-1533, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38530536

RESUMO

The primary goal of bone tissue engineering is to restore and rejuvenate bone defects by using a suitable three-dimensional scaffold, appropriate cells, and growth hormones. Various scaffolding methods are used to fabricate three-dimensional scaffolds, which provide the necessary environment for cell activity and bone formation. Multiple materials may be used to create scaffolds with hierarchical structures that are optimal for cell growth and specialization. This study examines a notion for creating an optimal framework for bone regeneration using a combination of the robocasting method and the electrospinning approach. Research indicates that the integration of these two procedures enhances the benefits of each method and provides a rationale for addressing their shortcomings via this combination. The hybrid approach is anticipated to provide a manufactured scaffold that can effectively replace bone defects while possessing the necessary qualities for bone regeneration.


Assuntos
Regeneração Óssea , Impressão Tridimensional , Engenharia Tecidual , Alicerces Teciduais , Alicerces Teciduais/química , Humanos , Engenharia Tecidual/métodos , Animais , Osso e Ossos , Osteogênese
14.
J Biomed Mater Res A ; 112(9): 1472-1483, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38477071

RESUMO

Critical-sized bone defects pose a significant challenge in advanced healthcare due to limited bone tissue regenerative capacity. The complex interplay of numerous overlapping variables hinders the development of multifunctional biocomposites. Phytochemicals show promise in promoting bone growth, but their dose-dependent nature and physicochemical properties halt clinical use. To develop a comprehensive solution, a 3D-printed (3DP) extrusion-based tricalcium phosphate-polycaprolactone (TCP-PCL) scaffold is augmented with quercetin and potassium chloride (KCl). This composite material demonstrates a compressive strength of 30 MPa showing promising stability for low load-bearing applications. Quercetin release from the scaffold follows a biphasic pattern that persists for up to 28 days, driven via diffusion-mediated kinetics. The incorporation of KCl allows for tunable degradation rates of scaffolds and prevents the initial rapid release. Functionalization of scaffolds facilitates the attachment and proliferation of human fetal osteoblasts (hfOB), resulting in a 2.1-fold increase in cell viability. Treated scaffolds exhibit a 3-fold reduction in osteosarcoma (MG-63) cell viability as compared to untreated substrates. Ruptured cell morphology and decreased mitochondrial membrane potential indicate the antitumorigenic potential. Scaffolds loaded with quercetin and quercetin-KCl (Q-KCl) demonstrate 76% and 89% reduction in bacterial colonies of Staphylococcus aureus, respectively. This study provides valuable insights as a promising strategy for bone tissue engineering (BTE) in orthopedic repair.


Assuntos
Fosfatos de Cálcio , Poliésteres , Impressão Tridimensional , Quercetina , Engenharia Tecidual , Alicerces Teciduais , Quercetina/farmacologia , Quercetina/química , Humanos , Poliésteres/química , Engenharia Tecidual/métodos , Fosfatos de Cálcio/química , Fosfatos de Cálcio/farmacologia , Alicerces Teciduais/química , Osso e Ossos/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoblastos/citologia , Sobrevivência Celular/efeitos dos fármacos , Linhagem Celular Tumoral
15.
Biomech Model Mechanobiol ; 23(3): 987-1012, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38416219

RESUMO

Recently, 3D-printed biodegradable scaffolds have shown great potential for bone repair in critical-size fractures. The differentiation of the cells on a scaffold is impacted among other factors by the surface deformation of the scaffold due to mechanical loading and the wall shear stresses imposed by the interstitial fluid flow. These factors are in turn significantly affected by the material properties, the geometry of the scaffold, as well as the loading and flow conditions. In this work, a numerical framework is proposed to study the influence of these factors on the expected osteochondral cell differentiation. The considered scaffold is rectangular with a 0/90 lay-down pattern and a four-layered strut made of polylactic acid with a 5% steel particle content. The distribution of the different types of cells on the scaffold surface is estimated through a scalar stimulus, calculated by using a mechanobioregulatory model. To reduce the simulation time for the computation of the stimulus, a probabilistic machine learning (ML)-based reduced-order model (ROM) is proposed. Then, a sensitivity analysis is performed using the Shapley additive explanations to examine the contribution of the various parameters to the framework stimulus predictions. In a final step, a multiobjective optimization procedure is implemented using genetic algorithms and the ROM, aiming to identify the material parameters and loading conditions that maximize the percentage of surface area populated by bone cells while minimizing the area corresponding to the other types of cells and the resorption condition. The results of the performed analysis highlight the potential of using ROMs for the scaffold design, by dramatically reducing the simulation time while enabling the efficient implementation of sensitivity analysis and optimization procedures.


Assuntos
Osso e Ossos , Aprendizado de Máquina , Engenharia Tecidual , Alicerces Teciduais , Alicerces Teciduais/química , Engenharia Tecidual/métodos , Osso e Ossos/fisiologia , Probabilidade , Estresse Mecânico , Humanos , Simulação por Computador , Poliésteres
16.
Acta Biomater ; 177: 506-524, 2024 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-38360290

RESUMO

The application of 3D printing to calcium phosphates has opened unprecedented possibilities for the fabrication of personalized bone grafts. However, their biocompatibility and bioactivity are counterbalanced by their high brittleness. In this work we aim at overcoming this problem by developing a self-hardening ink containing reactive ceramic particles in a polycaprolactone solution instead of the traditional approach that use hydrogels as binders. The presence of polycaprolactone preserved the printability of the ink and was compatible with the hydrolysis-based hardening process, despite the absence of water in the ink and its hydrophobicity. The microstructure evolved from a continuous polymeric phase with loose ceramic particles to a continuous network of hydroxyapatite nanocrystals intertwined with the polymer, in a configuration radically different from the polymer/ceramic composites obtained by fused deposition modelling. This resulted in the evolution from a ductile behavior, dominated by the polymer, to a stiffer behavior as the ceramic phase reacted. The polycaprolactone binder provides two highly relevant benefits compared to hydrogel-based inks. First, the handleability and elasticity of the as-printed scaffolds, together with the proven possibility of eliminating the solvent, opens the door to implanting the scaffolds freshly printed once lyophilized, while in a ductile state, and the hardening process to take place inside the body, as in the case of calcium phosphate cements. Second, even with a hydroxyapatite content of more than 92 wt.%, the flexural strength and toughness of the scaffolds after hardening are twice and five times those of the all-ceramic scaffolds obtained with the hydrogel-based inks, respectively. STATEMENT OF SIGNIFICANCE: Overcoming the brittleness of ceramic scaffolds would extend the applicability of synthetic bone grafts to high load-bearing situations. In this work we developed a 3D printing ink by replacing the conventional hydrogel binder with a water-free polycaprolactone solution. The presence of polycaprolactone not only enhanced significantly the strength and toughness of the scaffolds while keeping the proportion of bioactive ceramic phase larger than 90 wt.%, but it also conferred flexibility and manipulability to the as-printed scaffolds. Since they are able to harden upon contact with water under physiological conditions, this opens up the possibility of implanting them immediately after printing, while they are still in a ductile state, with clear advantages for fixation and press-fit in the bone defect.


Assuntos
Durapatita , Alicerces Teciduais , Alicerces Teciduais/química , Tinta , Biomimética , Poliésteres , Hidrogéis/química , Impressão Tridimensional , Água , Engenharia Tecidual
17.
Heliyon ; 10(4): e26005, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38375289

RESUMO

The present study investigated the influence of pore size of strut-based Diamond and surface-based Gyroid structures for their suitability as medical implants. Samples were made additively from laser powder bed fusion process with a relative density of 0.3 and pore sizes ranging from 300 to 1300 µm. They were subsequently examined for their manufacturability and mechanical properties. In addition, non-Newtonian computational fluid dynamics and discrete phase models were conducted to assess pressure drop and cell seeding efficiency. The results showed that both Diamond and Gyroid had higher as-built densities with smaller pore sizes. However, Gyroid demonstrated better manufacturability as its relative density was closer to the as-designed one. In addition, based on mechanical testing, the elastic modulus was largely unaffected by pore size, but post-yielding behaviors differed, especially in Diamond. High mechanical sensitivity in Diamond could be explained partly by Finite Element simulations, which revealed stress localization in Diamond and more uniform stress distribution in Gyroid. Furthermore, we defined the product of the normalized specific surface, normalized pressure drop, and cell seeding efficiency as the indicator of an optimal pore size, in which this factor identified an optimal pore size of approximately 500 µm for both Diamond and Gyroid. Besides, based on such criterion, Gyroid exhibited greater applicability as bone scaffolds. In summary, this study provides comprehensive assessment of the effect of pore size and demonstrates the efficient estimation of an in-silico framework for evaluating lattice structures as medical implants, which could be applied to other lattice architectures.

18.
Nanomaterials (Basel) ; 14(2)2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38251117

RESUMO

A large amount of research in orthopedic and maxillofacial domains is dedicated to the development of bioactive 3D scaffolds. This includes the search for highly resorbable compounds, capable of triggering cell activity and favoring bone regeneration. Considering the phosphocalcic nature of bone mineral, these aims can be achieved by the choice of amorphous calcium phosphates (ACPs). Because of their metastable property, these compounds are however to-date seldom used in bulk form. In this work, we used a non-conventional "cold sintering" approach based on ultrafast low-pressure RT compaction to successfully consolidate ACP pellets while preserving their amorphous nature (XRD). Complementary spectroscopic analyses (FTIR, Raman, solid-state NMR) and thermal analyses showed that the starting powder underwent slight physicochemical modifications, with a partial loss of water and local change in the HPO42- ion environment. The creation of an open porous structure, which is especially adapted for non-load bearing bone defects, was also observed. Moreover, the pellets obtained exhibited sufficient mechanical resistance allowing for manipulation, surgical placement and eventual cutting/reshaping in the operation room. Three-dimensional porous scaffolds of cold-sintered reactive ACP, fabricated through this low-energy, ultrafast consolidation process, show promise toward the development of highly bioactive and tailorable biomaterials for bone regeneration, also permitting combinations with various thermosensitive drugs.

19.
J Mech Behav Biomed Mater ; 150: 106258, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38000162

RESUMO

In bone tissue engineering (BTE), defects in large bones remain the greatest issue which can be addressed using bone scaffolds. In this work, blends of heat cured polymethylmethacrylate (HC PMMA) and various weight percentages of poly-ether-ketone-ketone (PEKK) (0, 2, 4, 8, and 10%) were made using a porogen leaching process. The blends were then subjected to tensile, compression and bending tests to select the optimum blend. Based on the results obtained, HC PMMA blended with 2 wt% PEKK was selected to produce the bio-porous blends. Here, the porosity was imparted using tartaric acid (C4H6O6) and sodium hydrogen carbonate (NaHCO3) as porogen leaching components. Porous blends resulted were then reinforced with a nano titanium dioxide powder (nTiO2) at different weight percentages of (0, 1, 3, and 5). The results showed that porous composites made of (HC PMMA: 2 wt% PEKK) blend reinforced with 5 wt % nTiO2 exhibit the highest strength values under various loadings. The FTIR identified the functional groups of the bone scaffold components. The mean pore size and pore depth were measured using atomic force microscopy (AFM) analysis and the values are 92.6 nm and 42.78 nm, respectively. The good distribution of the PEKK and nTiO2 within the HC PMMA and the uniform porous structure with multi-scale pores between 535 nm and 1.187 mm were confirmed by the AFM data and SEM images, respectively. This research expected that the porous composite (HC PMMA: 2% PEKK: 5% nTiO2) is a good candidate for bone scaffold applications.


Assuntos
Cetonas , Polimetil Metacrilato , Polimetil Metacrilato/química , Cetonas/química , Éter , Temperatura Alta , Engenharia Tecidual , Porosidade , Etil-Éteres , Éteres , Alicerces Teciduais/química
20.
Front Bioeng Biotechnol ; 11: 1305936, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38107615

RESUMO

Modern orthopaedic implants use lattice structures that act as 3D scaffolds to enhance bone growth into and around implants. Stochastic scaffolds are of particular interest as they mimic the architecture of trabecular bone and can combine isotropic properties and adjustable structure. The existing research mainly concentrates on controlling the mechanical and biological performance of periodic lattices by adjusting pore size and shape. Still, less is known on how we can control the performance of stochastic lattices through their design parameters: nodal connectivity, strut density and strut thickness. To elucidate this, four lattice structures were evaluated with varied strut densities and connectivity, hence different local geometry and mechanical properties: low apparent modulus, high apparent modulus, and two with near-identical modulus. Pre-osteoblast murine cells were seeded on scaffolds and cultured in vitro for 28 days. Cell adhesion, proliferation and differentiation were evaluated. Additionally, the expression levels of key osteogenic biomarkers were used to assess the effect of each design parameter on the quality of newly formed tissue. The main finding was that increasing connectivity increased the rate of osteoblast maturation, tissue formation and mineralisation. In detail, doubling the connectivity, over fixed strut density, increased collagen type-I by 140%, increased osteopontin by 130% and osteocalcin by 110%. This was attributed to the increased number of acute angles formed by the numerous connected struts, which facilitated the organization of cells and accelerated the cell cycle. Overall, increasing connectivity and adjusting strut density is a novel technique to design stochastic structures which combine a broad range of biomimetic properties and rapid ossification.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA