Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Adv Mater ; : e2400700, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38842622

ABSTRACT

The growing aging population, with its associated chronic diseases, underscores the urgency for effective tissue regeneration strategies. Biomaterials have played a pivotal role in the realm of tissue reconstruction and regeneration, with a distinct shift towards minimally invasive (MI) treatments. This transition, fueled by engineered biomaterials, has steered away from invasive surgical procedures to embrace approaches offering reduced trauma, accelerated recovery, and cost-effectiveness. In the realm of MI tissue repair and cargo delivery, various techniques have been explored. While in situ polymerization has been prominent, it is not without its challenges. This narrative review explores diverse biomaterials, fabrication methods, and biofunctionalization for injectable pre-formed scaffolds, focusing on their unique advantages. The injectable pre-formed scaffolds, exhibiting compressibility, controlled injection, and maintained mechanical integrity, emerge as promising alternative solutions to in situ polymerization challenges. The conclusion of this review emphasizes the importance of interdisciplinary design facilitated by synergizing fields of materials science, advanced 3D biomanufacturing, and mechanobiological studies, and innovative approaches for effective MI tissue regeneration. This article is protected by copyright. All rights reserved.

2.
Adv Healthc Mater ; 13(12): e2303772, 2024 May.
Article in English | MEDLINE | ID: mdl-38271276

ABSTRACT

3D stem cell spheroids have immense potential for various tissue engineering applications. However, current spheroid fabrication techniques encounter cell viability issues due to limited oxygen access for cells trapped within the core, as well as nonspecific differentiation issues due to the complicated environment following transplantation. In this study, functional 3D spheroids are developed using mesenchymal stem cells with 2D hetero-nanostructures (HNSs) composed of single-stranded DNA (ssDNA) binding carbon nanotubes (sdCNTs) and gelatin-bind black phosphorus nanosheets (gBPNSs). An osteogenic molecule, dexamethasone (DEX), is further loaded to fabricate an sdCNTgBP-DEX HNS. This approach aims to establish a multifunctional cell-inductive 3D spheroid with improved oxygen transportation through hollow nanotubes, stimulated stem cell growth by phosphate ions supplied from BP oxidation, in situ immunoregulation, and osteogenesis induction by DEX molecules after implantation. Initial transplantation of the 3D spheroids in rat calvarial bone defect shows in vivo macrophage shifts to an M2 phenotype, leading to a pro-healing microenvironment for regeneration. Prolonged implantation demonstrates outstanding in vivo neovascularization, osteointegration, and new bone regeneration. Therefore, these engineered 3D spheroids hold great promise for bone repair as they allow for stem cell delivery and provide immunoregulative and osteogenic signals within an all-in-one construct.


Subject(s)
Bone Regeneration , Mesenchymal Stem Cells , Nanotubes, Carbon , Osteogenesis , Spheroids, Cellular , Animals , Osteogenesis/drug effects , Spheroids, Cellular/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Rats , Bone Regeneration/drug effects , Nanotubes, Carbon/chemistry , Dexamethasone/chemistry , Dexamethasone/pharmacology , Rats, Sprague-Dawley , Nanostructures/chemistry , Tissue Engineering/methods , Male , DNA, Single-Stranded/chemistry , Phosphorus/chemistry , Gelatin/chemistry
3.
J Biomed Mater Res A ; 112(5): 672-684, 2024 05.
Article in English | MEDLINE | ID: mdl-37971074

ABSTRACT

Polycaprolactone fumarate (PCLF) is a cross-linkable PCL derivative extensively considered for tissue engineering applications. Although injection molding has been widely used to develop PCLF scaffolds, platforms developed using such technique lack precise control on architecture, design, and porosity required to ensure adequate cellular and tissue responses. In particular, the scaffolds should provide a suitable surface for cell attachment and proliferation, and facilitate cell-cell communication and nutrient flow. 3D printing technologies have led to new architype for biomaterial development with micro-architecture mimicking native tissue. Here, we developed a method for 3D printing of PCLF structures using the extrusion printing technique. The crosslinking property of PCLF enabled the unique post-processing of 3D printed scaffolds resulting in highly porous and flexible PCLF scaffolds with compressive properties imitating natural features of cancellous bone. Generated scaffolds supported excellent attachment and proliferation of mesenchymal stem cells (MSC). The high porosity of PCLF scaffolds facilitated vascularized membrane formation demonstrable with the stringency of the ex ovo chicken chorioallantoic membrane (CAM) implantation. Furthermore, upon implantation to rat calvarium defects, PCLF scaffolds enabled an exceptional new bone formation with a bone mineral density of newly formed bone mirroring native bone tissue. These studies suggest that the 3D-printed highly porous PCLF scaffolds may serve as a suitable biomaterial platform to significantly expand the utility of the PCLF biomaterial for bone tissue engineering applications.


Subject(s)
Fumarates , Tissue Scaffolds , Rats , Animals , Tissue Scaffolds/chemistry , Fumarates/pharmacology , Fumarates/chemistry , Biocompatible Materials/chemistry , Polyesters/pharmacology , Polyesters/chemistry , Tissue Engineering/methods , Bone Regeneration , Porosity , Printing, Three-Dimensional
4.
Biomater Adv ; 153: 213539, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37429047

ABSTRACT

Pre-formed hydrogel scaffolds have emerged as favorable vehicles for tissue regeneration, promoting minimally invasive treatment of native tissue. However, due to the high degree of swelling and inherently poor mechanical properties, development of complex structural hydrogel scaffolds at different dimensional scales has been a continuous challenge. Herein, we take a novel approach at the intersections of engineering design and bio-ink chemistry to develop injectable pre-formed structural hydrogel scaffolds fabricated via visible light (VL) induced digital light processing (DLP). In this study, we first determined the minimum concentration of poly(ethylene glycol) diacrylate (PEGDA) to be added to the gelatin methacrylate (GelMA) bio-ink in order to achieve scalable and high printing-fidelity with desired cell adhesion, viability, spreading, and osteogenic differentiation characteristics. Despite the advantages of hybrid GelMA-PEGDA bio-ink in improving scalability and printing-fidelity, compressibility, shape-recovery, and injectability of the 3D bioprinted scaffolds were compromised. To restore these needed characteristics for minimally invasive tissue regeneration applications, we performed topological optimization to design highly compressible and injectable pre-formed (i.e., 3D bioprinted) microarchitectural scaffolds. The designed injectable pre-formed microarchitectural scaffolds showed a great capacity to retain the viability of the encapsulated cells (>72 % after 10 cycles of injection). Lastly, ex ovo chicken chorioallantoic membrane (CAM) studies revealed that the optimized injectable pre-formed hybrid hydrogel scaffold is biocompatible and supports angiogenic growth.


Subject(s)
Osteogenesis , Tissue Scaffolds , Tissue Scaffolds/chemistry , Hydrogels , Light , Gelatin/chemistry
5.
ACS Biomater Sci Eng ; 9(3): 1585-1597, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36854041

ABSTRACT

Current bone cement systems often demand free radical or metal-related initiators and/or catalysts for the crosslinking process, which may cause serious toxicity to the human body. In addition, the resultant dense scaffolds may have a prolonged degradation time and are difficult for cells to infiltrate and form new tissue. In this study, we developed a porous "click" organic-inorganic nanohybrid (PO-click-ON) cement that crosslinks via metal-free biorthogonal click chemistry and forms porous structures mimicking the native bone tissue via particulate leaching. Strain-promoted click reaction enables fast and efficient crosslinking of polymer chains with the exclusion of any toxic initiator or catalyst. The resulting PO-click-ON implants supported exceptional in vitro stem cell adhesion and osteogenic differentiation with a large portion of stem cells infiltrated deep into the scaffolds. In vivo study using a rat cranial defect model demonstrated that the PO-click-ON system achieved outstanding cell adsorption, neovascularization, and bone formation. The porous click cement developed in this study serves as a promising platform with multifunctionality for bone and other tissue engineering applications.


Subject(s)
Bone Cements , Osteogenesis , Humans , Rats , Animals , Bone Cements/chemistry , Tissue Engineering/methods , Bone and Bones , Stem Cells
6.
J Mech Behav Biomed Mater ; 138: 105612, 2023 02.
Article in English | MEDLINE | ID: mdl-36509012

ABSTRACT

Functional cellular structures with controllable mechanical and morphological properties are of great interest for applications including tissue engineering, energy storage, and aerospace. Additive manufacturing (AM), also referred to as 3D printing, has enabled the potential for fabrication of functional porous scaffolds (i.e., meta-biomaterials) with controlled geometrical, morphological, and mechanical properties. Understanding the biomechanical behavior of 3D printed porous scaffolds under physiologically relevant loading and environmental conditions is crucial in accurately predicting the in vivo performance. This study was aimed to investigate the environmental dependency of the mechanical responses of 3D printed porous scaffolds of poly(methyl methacrylate) (PMMA) Class IIa biomaterial that was based on triply periodic minimal surfaces - TPMS (i.e., Primitive and Schoen-IWP). The 3D printed scaffolds (n = 5/study group) were tested under compressive loading in both ambient and fluidic (distilled water with pH = 7.4) environments according to ASTM D1621 standards. Outcomes of this study showed that compressive properties of the developed scaffolds are significantly lower in the fluidic condition than the ambient environment for the same topological and morphological group (p≤0.023). Additionally, compressive properties and flexural stiffness of the studied scaffolds were within the range of trabecular bone's properties, for both topological classes. Relationships between predicted mechanical responses and morphological properties (i.e., porosity) were evaluated for each topological class. Quantitative correlation analysis indicated that mechanical behavior of the developed 3D printed scaffolds can be controlled based on both topology and morphology.


Subject(s)
Biomimetics , Polymethyl Methacrylate , Biocompatible Materials/chemistry , Tissue Engineering , Porosity , Printing, Three-Dimensional , Tissue Scaffolds/chemistry
7.
Biomaterials ; 284: 121507, 2022 05.
Article in English | MEDLINE | ID: mdl-35421800

ABSTRACT

The nanomaterials research spectrum has seen the continuous emergence of two-dimensional (2D) materials over the years. These highly anisotropic and ultrathin materials have found special attention in developing biomedical platforms for therapeutic applications, biosensing, drug delivery, and regenerative medicine. Three-dimensional (3D) printing and bioprinting technologies have emerged as promising tools in medical applications. The convergence of 2D nanomaterials with 3D printing has extended the application dynamics of available biomaterials to 3D printable inks and bioinks. Furthermore, the unique properties of 2D nanomaterials have imparted multifunctionalities to 3D printed constructs applicable to several biomedical applications. 2D nanomaterials such as graphene and its derivatives have long been the interest of researchers working in this area. Beyond graphene, a range of emerging 2D nanomaterials, such as layered silicates, black phosphorus, transition metal dichalcogenides, transition metal oxides, hexagonal boron nitride, and MXenes, are being explored for the multitude of biomedical applications. Better understandings on both the local and systemic toxicity of these materials have also emerged over the years. This review focuses on state-of-art 3D fabrication and biofabrication of biomedical platforms facilitated by 2D nanomaterials, with the comprehensive summary of studies focusing on the toxicity of these materials. We highlight the dynamism added by 2D nanomaterials in the printing process and the functionality of printed constructs.


Subject(s)
Bioprinting , Graphite , Nanostructures , Bioprinting/methods , Oxides , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds
8.
Comput Biol Med ; 137: 104833, 2021 10.
Article in English | MEDLINE | ID: mdl-34534795

ABSTRACT

Osteoporosis-related vertebral compression fracture can occur under normal physiological activities. Bone metastasis is another source of vertebral fracture. Different loading rates, either high-energy traumas such as falls or low-energy traumas under normal physiological activities, can result in different fracture outcomes. The aim of the current study was to develop a quantitative computed tomography-based finite element analysis (QCT/FEA) technique for single vertebral bodies to predict fracture strength of three-level spine segments. Developed QCT/FEA technique was also used to characterize vertebral elastic moduli at two loading rates of 5 mm/min, representing a physiologic loading condition, and 12000 mm/min, representing a high-energy trauma. To this end, a cohort of human spine segments divided into three groups of intact, defect, and augmented were mechanically tested to fracture; then, experimental stiffness and fracture strength values were measured. Outcomes of this study showed no significant difference between the elastic modulus equations at the two testing speeds. Areal bone mineral density measured by dual x-ray absorptiometry (DXA/BMD) explained only 53% variability (R2 = 0.53) in fracture strength outcomes. However, QCT/FEA could explain 70% of the variability (R2 = 0.70) in experimentally measured fracture strength values. Adding disk degeneration grading, testing speed, and sex to QCT/FEA-estimated fracture strength values further increased the performance of our statistical model by 14% (adjusted R2 of 0.84 between the prediction and experimental fracture forces). In summary, our results indicated that a single-vertebra model, which is computationally less expensive and more time efficient, is capable of estimating fracture outcomes with acceptable performance (range: 70-84%).


Subject(s)
Fractures, Compression , Spinal Fractures , Absorptiometry, Photon , Bone Density , Finite Element Analysis , Fractures, Compression/diagnostic imaging , Humans , Spinal Fractures/diagnostic imaging , Spine
9.
PLoS One ; 16(7): e0253786, 2021.
Article in English | MEDLINE | ID: mdl-34260623

ABSTRACT

Design and processing capabilities of additive manufacturing (AM) to fabricate complex geometries continues to drive the adoption of AM for biomedical applications. In this study, a validated design methodology is presented to evaluate AM as an effective fabrication technique for reconstruction of large bone defects after tumor resection in pediatric oncology patients. Implanting off-the-shelf components in pediatric patients is especially challenging because most standard components are sized and shaped for more common adult cases. While currently reported efforts on AM implants are focused on maxillofacial, hip and knee reconstructions, there have been no reported studies on reconstruction of proximal humerus tumors. A case study of a 9-year-old diagnosed with proximal humerus osteosarcoma was used to develop a patient-specific AM prosthesis for the humerus following tumor resection. Commonly used body-centered cubic (BCC) structures were incorporated at the surgical neck and distal interface in order to increase the effective surface area, promote osseointegration, and reduce the implant weight. A patient-specific prosthesis was fabricated using electron beam melting method from biocompatible Ti-6Al-4V. Both computational and biomechanical tests were performed on the prosthesis to evaluate its biomechanical behavior under varying loading conditions. Morphological analysis of the construct using micro-computed tomography was used to compare the as-designed and as-built prosthesis. It was found that the patient-specific prosthesis could withstand physiologically-relevant loading conditions with minimal permanent deformation (82 µm after 105 cycles) at the medial aspect of the porous surgical neck. These outcomes support potential translation of the patient-specific AM prostheses to reconstruct large bone defects following tumor resection.


Subject(s)
Humerus/surgery , Osteosarcoma/surgery , Printing, Three-Dimensional , Prosthesis Design , Prosthesis Implantation/instrumentation , Child , Finite Element Analysis , Humans , Humerus/diagnostic imaging , Humerus/pathology , Imaging, Three-Dimensional , Materials Testing , Osseointegration , Osteosarcoma/pathology , Osteotomy/adverse effects , Porosity , Surface Properties , Titanium , X-Ray Microtomography
10.
Biomaterials ; 276: 121014, 2021 09.
Article in English | MEDLINE | ID: mdl-34280821

ABSTRACT

Injectable polymers have attracted intensive attention in tissue engineering and drug delivery applications. Current injectable polymer systems often require free-radical or heavy-metal initiators and catalysts for the crosslinking process, which may be extremely toxic to the human body. Here, we report a novel polyhedral oligomeric silsesquioxane (POSS) based strain-promoted alkyne-azide cycloaddition (SPAAC) "click" organic-inorganic nanohybrids (click-ON) system that can be click-crosslinked without any toxic initiators or catalysts. The click-ON scaffolds supported excellent adhesion, proliferation, and osteogenesis of stem cells. In vivo evaluation using a rat cranial defect model showed outstanding bone formation with minimum cytotoxicity. Essential osteogenic alkaline phosphatase (ALP) and vascular CD31 marker expression were detected on the defect site, indicating excellent support of in vivo osteogenesis and vascularization. Using salt leaching techniques, an injectable porous click-ON cement was developed to create porous structures and support better in vivo bone regeneration. Beyond defect filling, the click-ON cement also showed promising application for spinal fusion using rabbits as a model. Compared to the current clinically used poly (methyl methacrylate) (PMMA) cement, this click-ON cement showed great advantages of low heat generation, better biocompatibility and biodegradability, and thus has great potential for bone and related tissue engineering applications.


Subject(s)
Bone Cements , Tissue Engineering , Animals , Bone Regeneration , Hydrogels , Osteogenesis , Rabbits , Rats , Tissue Scaffolds
11.
J Mech Behav Biomed Mater ; 119: 104559, 2021 07.
Article in English | MEDLINE | ID: mdl-33915439

ABSTRACT

While several studies have investigated fracture outcomes of intact vertebrae, fracture properties in metastatically-involved and augmented vertebrae are still far from understood. Consequently, this study was aimed to use 3D digital image correlation (3D-DIC) method to investigate the failure properties of spine segments with simulated metastatic lesions, segments augmented with poly(propylene fumarate) (PPF), and compare the outcomes with intact spines. To this end, biomechanical experiments accompanied by 3D-DIC were performed on spine segments consisting of three vertebrae and two intervertebral discs (IVDs) at loading rates of 0.083 mm/s, mimicking a physiological loading condition, and 200 mm/s, mimicking an impact-type loading condition such as a fall or an accident. Full-field surface strain analysis indicated PPF augmentation reduces the superior/inferior strain when compared with the defect specimens; Presence of a defect in the middle vertebra resulted in shear band fracture pattern. Failure of the superior endplates was confirmed in several defect specimens as the superior IVDs were protruding out of defects. The augmenting PPF showed lower superior/inferior surface strain values at the fast speed as compared to the slow speed. The results of our study showed a significant increase in the fracture force from slow to fast speeds (p = 0.0246). The significance of the study was to determine the fracture properties of normal, pathological, and augmented spinal segments under physiologically-relevant loading conditions. Understanding failure properties associated with either defect (i.e., metastasis lesion) or augmented (i.e., post-treatment) spine segments could potentially provide new insights on the outcome prediction and treatment planning. Additionally, this study provides new knowledge on the effect of PPF augmentation in improving fracture properties, potentially decreasing the risk of fracture in osteoporotic and metastatic spines.


Subject(s)
Intervertebral Disc , Spinal Fractures , Biomechanical Phenomena , Cadaver , Humans , Lumbar Vertebrae , Spine
12.
Biomater Sci ; 9(8): 2768-2803, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33620047

ABSTRACT

Phosphorene, also known as black phosphorus (BP), is a two-dimensional (2D) material that has gained significant attention in several areas of current research. Its unique properties such as outstanding surface activity, an adjustable bandgap width, favorable on/off current ratios, infrared-light responsiveness, good biocompatibility, and fast biodegradation differentiate this material from other two-dimensional materials. The application of BP in the biomedical field has been rapidly emerging over the past few years. This article aimed to provide a comprehensive review of the recent progress on the unique properties and extensive medical applications for BP in bone, nerve, skin, kidney, cancer, and biosensing related treatment. The details of applications of BP in these fields were summarized and discussed.


Subject(s)
Nanotubes, Carbon , Neoplasms , Quantum Dots , Bone and Bones , Humans , Phosphorus
13.
J Biomech Eng ; 142(8)2020 08 01.
Article in English | MEDLINE | ID: mdl-31913444

ABSTRACT

Internal fixation with the use of locking plates is the standard surgical treatment for proximal humerus fractures, one of the most common fractures in the elderly. Screw cut-out through weak cancellous bone of the humeral head, which ultimately results in collapse of the fixed fracture, is the leading cause of failure and revision surgery. In an attempt to address this problem, surgeons often attach the plate with as many locking screws as possible into the proximal fragment. It is not thoroughly understood which screws and screw combinations play the most critical roles in fixation stability. This study conducted a detailed finite element analysis to evaluate critical parameters associated with screw cut-out failure. Several clinically relevant screw configurations and fracture gap sizes were modeled. Findings demonstrate that in perfectly reduced fracture cases, variation of the screw configurations had minor influence on mechanical stability of the fixation. The effects of screw configurations became substantial with the existence of a fracture gap. Interestingly, the use of a single anterior calcar screw was as effective as utilizing two screws to support the calcar. On the other hand, the variation in calcar screw configuration had minor influence on the fixation stability when all the proximal screws (A-D level) were filled. This study evaluates different screw configurations to further understand the influence of combined screw configurations and the individual screws on the fixation stability. Findings from this study may help decrease the risk for screw cut-out with proximal humerus varus collapse and the associated economic costs.


Subject(s)
Finite Element Analysis , Shoulder Fractures , Aged , Biomechanical Phenomena , Bone Plates , Fracture Fixation, Internal , Humans
14.
Ann Biomed Eng ; 48(1): 463-476, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31555983

ABSTRACT

Achieving satisfactory fracture fixation in osteoporotic patients with unstable proximal humerus fractures remains a major clinical challenge. Varus collapse is one of the more prominent complications that may lead to screw cutout. This aim of this study was to compare the fixation provided by conventional locking plates with novel design concepts that are only feasible through additive manufacturing (AM) techniques. In addition to reversed engineered implants, two novel implant designs with integrated struts were included in the study to provide medial support to humeral head. The medial strut was either solid or included a porous lattice structure intended to promote bone ingrowth. Biomechanical tests were performed using low density synthetic bones with simulated 3-part comminuted fractures. Nondestructive torsion and compression were performed, followed by increasing cyclic loading. The relative displacements between the bone fragments were determined using a 3D motion capture system. The AM manufactured implants with medial strut showed significant reduction of varus displacement during the increasing cyclic loading when compared to conventional designs. AM reversed-engineered locking plates showed similar mechanical behavior to conventional plates with identical geometry. This study demonstrates the feasibility and potential of employing alternative design via AM for fixation of unstable comminuted proximal humerus fractures to reduce fragment displacement.


Subject(s)
Bone Plates , Humerus , Prostheses and Implants , Shoulder Fractures/therapy , Biomechanical Phenomena , Fracture Fixation
SELECTION OF CITATIONS
SEARCH DETAIL
...