Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
Add more filters

Publication year range
1.
J Nanobiotechnology ; 22(1): 500, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169401

ABSTRACT

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.


Subject(s)
Bioprinting , Bone and Bones , Organoids , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Humans , Tissue Engineering/methods , Organoids/cytology , Bioprinting/methods , Animals , Bone Regeneration , Bone Transplantation/methods
2.
Bioprocess Biosyst Eng ; 47(9): 1483-1498, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38869621

ABSTRACT

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.


Subject(s)
Porifera , Printing, Three-Dimensional , Tissue Scaffolds , Animals , Tissue Scaffolds/chemistry , Porifera/chemistry , Mice , Silicon Dioxide/chemistry , Bone Regeneration , Porosity , Cell Survival , Tissue Engineering/methods , Cell Line , Bone and Bones
3.
Int J Mol Sci ; 24(4)2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36835168

ABSTRACT

Synthetic bone substitute materials (BSMs) are becoming the general trend, replacing autologous grafting for bone tissue engineering (BTE) in orthopedic research and clinical practice. As the main component of bone matrix, collagen type I has played a critical role in the construction of ideal synthetic BSMs for decades. Significant strides have been made in the field of collagen research, including the exploration of various collagen types, structures, and sources, the optimization of preparation techniques, modification technologies, and the manufacture of various collagen-based materials. However, the poor mechanical properties, fast degradation, and lack of osteoconductive activity of collagen-based materials caused inefficient bone replacement and limited their translation into clinical reality. In the area of BTE, so far, attempts have focused on the preparation of collagen-based biomimetic BSMs, along with other inorganic materials and bioactive substances. By reviewing the approved products on the market, this manuscript updates the latest applications of collagen-based materials in bone regeneration and highlights the potential for further development in the field of BTE over the next ten years.


Subject(s)
Biomimetic Materials , Bone Substitutes , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Bone and Bones , Collagen/chemistry , Biomimetic Materials/chemistry , Bone Regeneration , Bone Substitutes/chemistry , Biocompatible Materials/chemistry
4.
J Biol Regul Homeost Agents ; 33(6 Suppl. 2): 143-154 DENTAL SUPPLEMENT, 2019.
Article in English | MEDLINE | ID: mdl-32425035

ABSTRACT

Osseo-degeneration is a disorder related to several factors, that may lead to the disruption of several skeletal regions providing support, such as the femur head, the vertebrae and the alveolar bone. The functional condition can be restored by means of grafting procedures, using different materials: calcium powder, xenografts, ceramics and metals. Such procedures aim at reforming an adequate bone volume and strength, that is necessary to support loading forces. Bone regeneration requires that the basic biological principles of osteogenesis, osteoinduction, osteoconduction and biocompatibility are followed. The success of regenerative procedures may depend on the inner structural, mechanical and metabolic condition of the host's bone on which implants should be inserted, on the surgical technique, and on the biomaterial used. Among these, the aging process of the patient appears to be relevant. It can be associated with metabolic disease leading to systemic functional decay, which involves a gradual steady decline of hormonal, immune function and osteo-metabolic activity. The latter can affect the positive outcomes of bone reconstruction and implant therapy. This review will analyze the biological and physiological factors involved in the bone tissue break-down, such as the influences from gut microbiome unbalance and the consequent metabolic, endocrine, immune dysfunctions, the surgery procedures and the quality of the grafting material used. The decline of bone architecture and strength should be corrected by using an appropriate clinical regenerative approach, based on a bio-endocrine, metabolic and immunologic know-how. The final characteristics of the regenerated bone must be able to support the loading forces transmitted by the implants, independent of the body location, and should be individualized according to the different condition of each patient.


Subject(s)
Bone Diseases/therapy , Bone Substitutes , Bone Regeneration , Bone Transplantation , Bone and Bones , Ceramics , Gastrointestinal Microbiome , Humans , Osteogenesis
5.
Adv Exp Med Biol ; 881: 79-94, 2015.
Article in English | MEDLINE | ID: mdl-26545745

ABSTRACT

Survival of functional tissue constructs of clinically relevant size depends on the formation of an organized and uniformly distributed network of blood vessels and capillaries. The lack of such vasculature leads to spatio-temporal gradients in oxygen, nutrients and accumulation of waste products inside engineered tissue constructs resulting in negative biological events at the core of the scaffold. Unavailability of a well-defined vasculature also results in ineffective integration of scaffolds to the host vasculature upon implantation. Arguably, one of the greatest challenges in engineering clinically relevant bone substitutes, therefore, has been the development of vascularized bone scaffolds. Various approaches ranging from peptide and growth factor functionalized biomaterials to hyper-porous scaffolds have been proposed to address this problem with reasonable success. An emerging alternative to address this challenge has been the fabrication of pre-vascularized scaffolds by taking advantage of biomanufacturing techniques, such as soft- and photo-lithography or 3D bioprinting, and cell-based approaches, where functional capillaries are engineered in cell-laden scaffolds prior to implantation. These strategies seek to engineer pre-vascularized tissues in vitro, allowing for improved anastomosis with the host vasculature upon implantation, while also improving cell viability and tissue development in vitro. This book chapter provides an overview of recent methods to engineer pre-vascularized scaffolds for bone regeneration. We first review the development of functional blood capillaries in bony structures and discuss controlled delivery of growth factors, co-culture systems, and on-chip studies to engineer vascularized cell-laden biomaterials. Lastly, we review recent studies using microfabrication techniques and 3D printing to engineer pre-vascularized scaffolds for bone tissue engineering.


Subject(s)
Bone Regeneration/physiology , Bone and Bones/physiology , Neovascularization, Physiologic/physiology , Tissue Engineering/methods , Tissue Scaffolds , Biocompatible Materials/metabolism , Bone and Bones/blood supply , Bone and Bones/cytology , Coculture Techniques/methods , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Tissue Engineering/trends
6.
Adv Exp Med Biol ; 881: 57-78, 2015.
Article in English | MEDLINE | ID: mdl-26545744

ABSTRACT

Over the past decades, there has been a substantial amount of innovation and research into tissue engineering and regenerative approaches for the craniofacial region. This highly complex area presents many unique challenges for tissue engineers. Recent research indicates that various forms of implantable biodegradable scaffolds may play a beneficial role in the clinical treatment of craniofacial pathological conditions. Additionally, the direct delivery of bioactive molecules may further increase de novo bone formation. While these strategies offer an exciting glimpse into potential future treatments, there are several challenges that still must be overcome. In this chapter, we will highlight both current surgical approaches for craniofacial reconstruction and recent advances within the field of bone tissue engineering. The clinical challenges and limitations of these strategies will help contextualize and inform future craniofacial tissue engineering strategies.


Subject(s)
Bone Substitutes/metabolism , Oral Surgical Procedures/methods , Tissue Engineering/methods , Tissue Scaffolds , Bone Diseases/physiopathology , Bone Diseases/surgery , Bone Regeneration/drug effects , Bone Regeneration/physiology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Maxillofacial Abnormalities/physiopathology , Maxillofacial Abnormalities/surgery , Oral Surgical Procedures/trends , Osteogenesis/drug effects , Osteogenesis/physiology , Regenerative Medicine/methods , Regenerative Medicine/trends , Tissue Engineering/trends
7.
Adv Sci (Weinh) ; 11(35): e2401589, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39018263

ABSTRACT

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.


Subject(s)
Bone Regeneration , Periosteum , Polylactic Acid-Polyglycolic Acid Copolymer , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Bone Regeneration/physiology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Porosity , Tissue Engineering/methods , Osteogenesis/physiology , Cell Differentiation , Humans , Disease Models, Animal , Biocompatible Materials/chemistry , Mice
8.
Front Bioeng Biotechnol ; 12: 1372636, 2024.
Article in English | MEDLINE | ID: mdl-38707506

ABSTRACT

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.

9.
Acta Biomater ; 177: 506-524, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38360290

ABSTRACT

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.


Subject(s)
Durapatite , Tissue Scaffolds , Tissue Scaffolds/chemistry , Ink , Biomimetics , Polyesters , Hydrogels/chemistry , Printing, Three-Dimensional , Water , Tissue Engineering
10.
J Biomed Mater Res A ; 112(9): 1472-1483, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38477071

ABSTRACT

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.


Subject(s)
Calcium Phosphates , Polyesters , Printing, Three-Dimensional , Quercetin , Tissue Engineering , Tissue Scaffolds , Quercetin/pharmacology , Quercetin/chemistry , Humans , Polyesters/chemistry , Tissue Engineering/methods , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Tissue Scaffolds/chemistry , Bone and Bones/drug effects , Osteoblasts/drug effects , Osteoblasts/cytology , Cell Survival/drug effects , Cell Line, Tumor
11.
Article in English | MEDLINE | ID: mdl-38943424

ABSTRACT

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.

12.
3D Print Addit Manuf ; 11(4): 1418-1440, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39360139

ABSTRACT

Bone is a complex connective tissue that serves as mechanical and structural support for the human body. Bones' fractures are common, and the healing process is physiologically complex and involves both mechanical and biological aspects. Tissue engineering of bone scaffolds holds great promise for the future treatment of bone injuries. However, conventional technologies to prepare bone scaffolds cannot provide the required properties of human bones. Over the past decade, three-dimensional (3D) printing or additive manufacturing technologies have enabled control over the creation of bone scaffolds with personalized geometries, appropriate materials, and tailored pores. This article aims to review recent advances in the fabrication of bone scaffolds for bone repair and regeneration. A detailed review of bone fracture repair and an in-depth discussion on conventional manufacturing and 3D printing techniques are introduced with an emphasis on novel studies concepts, potentials, and limitations.

13.
J Biomed Mater Res A ; 112(12): 2042-2054, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38864151

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents , Bone Substitutes , Metal Nanoparticles , Silver , Animals , Silver/chemistry , Silver/pharmacology , Rabbits , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Metal Nanoparticles/chemistry , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Cattle , Osteoblasts/drug effects , Osteoblasts/cytology , Hydrogels/chemistry , Hydrogels/pharmacology
14.
Nanomaterials (Basel) ; 14(2)2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38251117

ABSTRACT

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.

15.
J Mech Behav Biomed Mater ; 150: 106258, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38000162

ABSTRACT

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.


Subject(s)
Ketones , Polymethyl Methacrylate , Polymethyl Methacrylate/chemistry , Ketones/chemistry , Ether , Hot Temperature , Tissue Engineering , Porosity , Ethyl Ethers , Ethers , Tissue Scaffolds/chemistry
16.
Ann Biomed Eng ; 52(6): 1518-1533, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38530536

ABSTRACT

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.


Subject(s)
Bone Regeneration , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Humans , Tissue Engineering/methods , Animals , Bone and Bones , Osteogenesis
17.
Acta Biomater ; 180: 115-127, 2024 05.
Article in English | MEDLINE | ID: mdl-38642786

ABSTRACT

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.


Subject(s)
Ceramics , Osteogenesis , Tissue Scaffolds , Printing, Three-Dimensional , Ceramics/chemistry , Tissue Scaffolds/chemistry , Tissue Scaffolds/standards , Osteogenesis/physiology , Animals , Rabbits , Male , Surface Properties
18.
Int J Biol Macromol ; 279(Pt 2): 135182, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39216566

ABSTRACT

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.


Subject(s)
Bone Regeneration , Mannans , Nanoparticles , Neovascularization, Physiologic , Osteogenesis , Silicon Dioxide , Tissue Scaffolds , Animals , Silicon Dioxide/chemistry , Tissue Scaffolds/chemistry , Mannans/chemistry , Mannans/pharmacology , Osteogenesis/drug effects , Neovascularization, Physiologic/drug effects , Rats , Nanoparticles/chemistry , Porosity , Bone Regeneration/drug effects , Rats, Wistar , Nanocomposites/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Male , Tissue Engineering/methods , Angiogenesis
19.
Int J Nanomedicine ; 19: 8309-8336, 2024.
Article in English | MEDLINE | ID: mdl-39161358

ABSTRACT

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.


Subject(s)
Bone Regeneration , Copper , Graphite , Hydrogels , Rats, Sprague-Dawley , Skull , Tissue Engineering , Tissue Scaffolds , Animals , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Copper/chemistry , Copper/pharmacology , Graphite/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Skull/drug effects , Skull/injuries , Rats , Mice , Tissue Engineering/methods , Osteogenesis/drug effects , Mice, Inbred BALB C , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Male , Metal Nanoparticles/chemistry , Nanostructures/chemistry , Gelatin/chemistry , RAW 264.7 Cells
20.
Drug Deliv ; 31(1): 2391001, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39239763

ABSTRACT

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.


Subject(s)
Bone Neoplasms , Drug Delivery Systems , Osteosarcoma , Tissue Scaffolds , Osteosarcoma/drug therapy , Humans , Drug Delivery Systems/methods , Bone Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Bone and Bones/drug effects , Animals
SELECTION OF CITATIONS
SEARCH DETAIL