Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 19: 2655-2673, 2024.
Article in English | MEDLINE | ID: mdl-38500680

ABSTRACT

Introduction: Immunotherapy has revolutionized cancer treatment by harnessing the immune system to enhance antitumor responses while minimizing off-target effects. Among the promising cancer-specific therapies, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted significant attention. Methods: Here, we developed an ionizable lipid nanoparticle (LNP) platform to deliver TRAIL mRNA (LNP-TRAIL) directly to the tumor microenvironment (TME) to induce tumor cell death. Our LNP-TRAIL was formulated via microfluidic mixing and the induction of tumor cell death was assessed in vitro. Next, we investigated the ability of LNP-TRAIL to inhibit colon cancer progression in vivo in combination with a TME normalization approach using Losartan (Los) or angiotensin 1-7 (Ang(1-7)) to reduce vascular compression and deposition of extracellular matrix in mice. Results: Our results demonstrated that LNP-TRAIL induced tumor cell death in vitro and effectively inhibited colon cancer progression in vivo, particularly when combined with TME normalization induced by treatment Los or Ang(1-7). In addition, potent tumor cell death as well as enhanced apoptosis and necrosis was found in the tumor tissue of a group treated with LNP-TRAIL combined with TME normalization. Discussion: Together, our data demonstrate the potential of the LNP to deliver TRAIL mRNA to the TME and to induce tumor cell death, especially when combined with TME normalization. Therefore, these findings provide important insights for the development of novel therapeutic strategies for the immunotherapy of solid tumors.


Subject(s)
Colonic Neoplasms , Liposomes , Nanoparticles , Tumor Microenvironment , Animals , Mice , Ligands , Apoptosis , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Tumor Necrosis Factor-alpha , TNF-Related Apoptosis-Inducing Ligand/metabolism
3.
Int J Nanomedicine ; 18: 6153-6183, 2023.
Article in English | MEDLINE | ID: mdl-37915750

ABSTRACT

Carbon-based nanomaterials (CBNs) are a category of nanomaterials with various systems based on combinations of sp2 and sp3 hybridized carbon bonds, morphologies, and functional groups. CBNs can exhibit distinguished properties such as high mechanical strength, chemical stability, high electrical conductivity, and biocompatibility. These desirable physicochemical properties have triggered their uses in many fields, including biomedical applications. In this review, we specifically focus on applying CBNs as scaffolds in tissue engineering, a therapeutic approach whereby CBNs can act for the regeneration or replacement of damaged tissue. Here, an overview of the structures and properties of different CBNs will first be provided. We will then discuss state-of-the-art advancements of CBNs and hydrogels as scaffolds for regenerating various types of human tissues. Finally, a perspective of future potentials and challenges in this field will be presented. Since this is a very rapidly growing field, we expect that this review will promote interdisciplinary efforts in developing effective tissue regeneration scaffolds for clinical applications.


Subject(s)
Nanostructures , Tissue Engineering , Humans , Hydrogels/chemistry , Carbon , Tissue Scaffolds/chemistry
4.
Int J Nanomedicine ; 18: 5891-5904, 2023.
Article in English | MEDLINE | ID: mdl-37873551

ABSTRACT

Introduction: Chimeric antigen receptor (CAR) cell therapy represents a hallmark in cancer immunotherapy, with significant clinical results in the treatment of hematological tumors. However, current approved methods to engineer T cells to express CAR use viral vectors, which are integrative and have been associated with severe adverse effects due to constitutive expression of CAR. In this context, non-viral vectors such as ionizable lipid nanoparticles (LNPs) arise as an alternative to engineer CAR T cells with transient expression of CAR. Methods: Here, we formulated a mini-library of LNPs to deliver pDNA to T cells by varying the molar ratios of excipient lipids in each formulation. LNPs were characterized and screened in vitro using a T cell line (Jurkat). The optimized formulation was used ex vivo to engineer T cells derived from human peripheral blood mononuclear cells (PBMCs) for the expression of an anti-CD19 CAR (CAR-CD19BBz). The effectiveness of these CAR T cells was assessed in vitro against Raji (CD19+) cells. Results: LNPs formulated with different molar ratios of excipient lipids efficiently delivered pDNA to Jurkat cells with low cytotoxicity compared to conventional transfection methods, such as electroporation and lipofectamine. We show that CAR-CD19BBz expression in T cells was transient after transfection with LNPs. Jurkat cells transfected with our top-performing LNPs underwent activation when exposed to CD19+ target cells. Using our top-performing LNP-9-CAR, we were able to engineer human primary T cells to express CAR-CD19BBz, which elicited significant specific killing of CD19+ target cells in vitro. Conclusion: Collectively, our results show that LNP-mediated delivery of pDNA is a suitable method to engineer human T cells to express CAR, which holds promise for improving the production methods and broader application of this therapy in the future.


Subject(s)
Excipients , Nanoparticles , Humans , Leukocytes, Mononuclear , Plasmids/genetics , DNA/genetics , Lipids
5.
J Funct Biomater ; 14(9)2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37754851

ABSTRACT

The use of bioactive materials, such as Ximenia americana L., to stimulate the bone repair process has already been studied; however, the synergistic effects of its association with light emitting diode (LED) have not been reported. The present work aims to evaluate the effect of its stem bark extract incorporated into methacrylate gelatin hydrogel (GelMA) on the bone repair process using pure hydrogel and hydrogel associated with LED therapy. For this purpose, the GelMA hydrogel loaded with Ximenia americana L. extract (steam bark) was produced, characterized and applied in animal experiments. The tests were performed using 50 male Wistar rats (divided into 5 groups) submitted to an induced tibia diaphyseal fracture. The therapy effects were verified for a period of 15 and 30 days of treatment using histological analysis and Raman spectroscopy. After 15 days of induced lesion/treatment, the new bone formation was significantly higher in the GXG (GelMA + X. americana L.) group compared to the control group (p < 0.0001). After 30 days, a statistically significant difference was observed when comparing the GXLEDG (GelMA + X. americana L. + LED) and the control group (p < 0.0001), the GXG and the control group (p < 0.001), and when comparing the GG, GXG (p < 0.005) and GXLEDG (p < 0.001) groups. The results shows that the Ximenia americana L. stem extract incorporated into GelMA hydrogel associated with LED therapy is a potentiator for animal bone repair.

6.
iScience ; 26(2): 106039, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36761021

ABSTRACT

Three-dimensional (3D) bioprinting has emerged as a class of promising techniques in biomedical research for a wide range of related applications. Specifically, stereolithography apparatus (SLA) and digital light processing (DLP)-based vat-polymerization techniques are highly effective methods of bioprinting, which can be used to produce high-resolution and architecturally sophisticated structures. Our review aims to provide an overview of SLA- and DLP-based 3D bioprinting strategies, starting from factors that affect these bioprinting processes. In addition, we summarize the advances in bioinks used in SLA and DLP, including naturally derived and synthetic bioinks. Finally, the biomedical applications of both SLA- and DLP-based bioprinting are discussed, primarily centered on regenerative medicine and tissue modeling engineering.

7.
Mol Aspects Med ; 91: 101108, 2023 06.
Article in English | MEDLINE | ID: mdl-35987701

ABSTRACT

The field of precision medicine allows for tailor-made treatments specific to a patient and thereby improve the efficiency and accuracy of disease prevention, diagnosis, and treatment and at the same time would reduce the cost, redundant treatment, and side effects of current treatments. Here, the combination of organ-on-a-chip and bioprinting into engineering high-content in vitro tissue models is envisioned to address some precision medicine challenges. This strategy could be employed to tackle the current coronavirus disease 2019 (COVID-19), which has made a significant impact and paradigm shift in our society. Nevertheless, despite that vaccines against COVID-19 have been successfully developed and vaccination programs are already being deployed worldwide, it will likely require some time before it is available to everyone. Furthermore, there are still some uncertainties and lack of a full understanding of the virus as demonstrated in the high number new mutations arising worldwide and reinfections of already vaccinated individuals. To this end, efficient diagnostic tools and treatments are still urgently needed. In this context, the convergence of bioprinting and organ-on-a-chip technologies, either used alone or in combination, could possibly function as a prominent tool in addressing the current pandemic. This could enable facile advances of important tools, diagnostics, and better physiologically representative in vitro models specific to individuals allowing for faster and more accurate screening of therapeutics evaluating their efficacy and toxicity. This review will cover such technological advances and highlight what is needed for the field to mature for tackling the various needs for current and future pandemics as well as their relevancy towards precision medicine.


Subject(s)
COVID-19 , Humans , COVID-19 Vaccines , SARS-CoV-2 , Precision Medicine
8.
J Funct Biomater ; 13(4)2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36412893

ABSTRACT

Three-dimensional bioprinting is a powerful technique for manufacturing improved engineered tissues. Three-dimensional bioprinted hydrogels have significantly advanced the medical field to repair cartilage tissue, allowing for such constructs to be loaded with different components, such as cells, nanoparticles, and/or drugs. Cartilage, as an avascular tissue, presents extreme difficulty in self-repair when it has been damaged. In this way, hydrogels with optimal chemical and physical properties have been researched to respond to external stimuli and release various bioactive agents to further promote a desired tissue response. For instance, methacryloyl gelatin (GelMA) is a type of modified hydrogel that allows for the encapsulation of cells, as well as oxygen-releasing nanoparticles that, in the presence of an aqueous medium and through controlled porosity and swelling, allow for internal and external environmental exchanges. This review explores the 3D bioprinting of hydrogels, with a particular focus on GelMA hydrogels, to repair cartilage tissue. Recent advances and future perspectives are described.

9.
Polymers (Basel) ; 14(20)2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36297865

ABSTRACT

In this work, Coca-Cola® bottles were reused as a PET polymer (rPET) source to produce electrospun polymeric nanofibers. The nanofibers were electrospun from polymer solutions with different concentrations of reduced graphene oxide (rGO) incorporated for applications in somatosensory electrical stimulation. The rPET/rGO nanofiber mats were characterized by SEM, TEM, Raman, DSC, TGA, and DMA and the results showed that the incorporation of rGO in electrospun rPET fibers produced rPET/rGO composites. The rPET/rGO composites were then evaluated for possible application as dry electrodes. Moreover, with a preliminary test of numerous volunteers, the rPET/rGO dry electrode showed promising results. The rPET/rGO electrodes showed good performance and applicability to make dry electrodes, and these have applications as dry or wearable electrodes to produce electrochemical sensors.

10.
Int J Nanomedicine ; 17: 1111-1124, 2022.
Article in English | MEDLINE | ID: mdl-35309966

ABSTRACT

Introduction: Three of the main requirements that remain major challenges in tissue engineering of the knee meniscus are to engineer scaffolds with compatible anatomical shape, good mechanical properties, and microstructure able to mimic the architecture of the extracellular matrix (ECM). In this context, we presented a new biofabrication strategy to develop a three-dimensional (3D) meniscus-regenerative scaffold with custom-made macroscopic size and microarchitecture bioinspired by the organization of structural fibers of native tissue ECM. Methods: The concept was based on the combination of bioprinted cell-laden hydrogel (type 1 collagen) reinforced by multilayers of biomimetically aligned electrospun nanofibrous mats (polycaprolactone/carbon nanotubes, PCL/CNT), using a patient-specific 3D digital meniscus model reconstructed from MRI data by free and open-source software. Results: The results showed that the incorporation of aligned nanofibers sheets between the hydrogel layers enhanced the scaffold's structural integrity and shape fidelity compared to the nanofiber-free collagen hydrogel. Furthermore, mechanical compression tests demonstrated that the presence of nanofiber layers significantly improved the mechanical properties of the bioprinted construct. Importantly, the introduction of PCL/CNT nanofibrous mats between the layers of the bioprinted collagen hydrogel did not negatively affect cell viability, in which mesenchymal stem cells remained viable even after 7 days of culture within the scaffold. Conclusion: Overall, these findings evidence that this bioengineering approach offers a promising strategy for fabricating biomimetic meniscus scaffolds for tissue engineering.


Subject(s)
Meniscus , Nanofibers , Nanotubes, Carbon , Humans , Nanofibers/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry
11.
Bioengineering (Basel) ; 8(11)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34821717

ABSTRACT

Nanocomposite scaffolds based on the combination of polymeric nanofibers with nanohydroxyapatite are a promising approach within tissue engineering. With this strategy, it is possible to synthesize nanobiomaterials that combine the well-known benefits and advantages of polymer-based nanofibers with the osteointegrative, osteoinductive, and osteoconductive properties of nanohydroxyapatite, generating scaffolds with great potential for applications in regenerative medicine, especially as support for bone growth and regeneration. However, as efficiently incorporating nanohydroxyapatite into polymeric nanofibers is still a challenge, new methodologies have emerged for this purpose, such as electrodeposition, a fast, low-cost, adjustable, and reproducible technique capable of depositing coatings of nanohydroxyapatite on the outside of fibers, to improve scaffold bioactivity and cell-biomaterial interactions. In this short review paper, we provide an overview of the electrodeposition method, as well as a detailed discussion about the process of electrodepositing nanohydroxyapatite on the surface of polymer electrospun nanofibers. In addition, we present the main findings of the recent applications of polymeric micro/nanofibrous scaffolds coated with electrodeposited nanohydroxyapatite in tissue engineering. In conclusion, comments are provided about the future direction of nanohydroxyapatite electrodeposition onto polymeric nanofibers.

12.
J Funct Biomater ; 12(2)2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34200191

ABSTRACT

BACKGROUND: Several studies proved that anodic oxidation improves osseointegration. This study aimed to optimize osseointegration through anodization in dental implants, obtaining anatase phase and controlled nanotopography. METHODS: The division of the groups with 60 titanium implants was: control (CG); sandblasted (SG); anodized (AG): anodized pulsed current (duty cycle 30%, 30 V, 0.2 A and 1000 Hz). Before surgery, surface characterization was performed using Atomic Force Microscopy (AFM), Scanning Electron Microscopy (SEM), X-ray Dispersive Energy Spectroscopy (EDS) and Raman Spectroscopy. For in vivo tests, 10 New Zealand white rabbits received an implant from each group. The sacrifice period was 2 and 6 weeks (n = 5) and the specimens were subjected to computed microtomography (µCT) and reverse torque test. RESULTS: AFM and SEM demonstrated a particular nanotopography on the surface in AG; the anatase phase was proved by Raman spectroscopy. In the µCT and in the reverse torque test, the AG group presented better results than the other groups. CONCLUSION: The chemical composition and structure of the TiO2 film were positively affected by the anodizing technique, intensifying the biological characteristics in osseointegration.

13.
J Funct Biomater ; 12(2)2021 May 02.
Article in English | MEDLINE | ID: mdl-34063270

ABSTRACT

Different strategies have been employed to provide adequate nutrients for engineered living tissues. These have mainly revolved around providing oxygen to alleviate the effects of chronic hypoxia or anoxia that result in necrosis or weak neovascularization, leading to failure of artificial tissue implants and hence poor clinical outcome. While different biomaterials have been used as oxygen generators for in vitro as well as in vivo applications, certain problems have hampered their wide application. Among these are the generation and the rate at which oxygen is produced together with the production of the reaction intermediates in the form of reactive oxygen species (ROS). Both these factors can be detrimental for cell survival and can severely affect the outcome of such studies. Here we present calcium peroxide (CPO) encapsulated in polycaprolactone as oxygen releasing microparticles (OMPs). While CPO releases oxygen upon hydrolysis, PCL encapsulation ensures that hydrolysis takes place slowly, thereby sustaining prolonged release of oxygen without the stress the bulk release can endow on the encapsulated cells. We used gelatin methacryloyl (GelMA) hydrogels containing these OMPs to stimulate survival and proliferation of encapsulated skeletal myoblasts and optimized the OMP concentration for sustained oxygen delivery over more than a week. The oxygen releasing and delivery platform described in this study opens up opportunities for cell-based therapeutic approaches to treat diseases resulting from ischemic conditions and enhance survival of implants under severe hypoxic conditions for successful clinical translation.

14.
Mater Sci Eng C Mater Biol Appl ; 120: 111776, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33545906

ABSTRACT

A biomineralization processes is disclosed for engineering nanomaterials that support bone repair. The material was fabricated through a hot press process using electrospun poly(lactic acid) (PLA) matrix covered with hybrid composites of carbon nanotubes/graphene nanoribbons (GNR) and nanohydroxyapatite (nHA). Various scaffolds were devised [nHA/PLA, PLA/GNR, and PLA/nHA/GNR (1 and 3%)] and their structure and morphology characterized through Scanning electron microscopy (SEM), Energy dispersive X-ray spectroscopy (EDS), and Atomic force microscope (AFM). Moreover, thorough biocompatibility and toxicity studies were performed. Here, in vivo studies on toxicity and cytotoxicity were conducted in aqueous dispersions of the biomaterials at concentrations of 30, 60, and 120 µg/mL using the Allium cepa test. Further toxicity studies were performed through hemolysis toxicity tests and genotoxicity tests evaluating the damage index and damage frequencies of DNAs through comet assays with samples of the animals' peripheral blood, marrow, and liver. Additionally, the regenerative activity of the scaffolds was analyzed by measuring the cortical tibiae of rats oophorectomized implanted with the biomaterials. Biochemical analyzes [glutamic pyruvic transaminase (GPT), glutamic oxaloacetic transaminase (GOT), urea, calcium, phosphorus, and alkaline phosphatase (ALP)] were also performed on blood samples. The results suggested a toxicity and cytotoxicity level for the GNR biomaterials at a concentration of 60 and 120 µg/mL, but non-toxicity and cytotoxicity for the 30 µg/mL concentration. The scaffolds obtained at a concentration of 0.3 mg/cm2 were not toxic in the hemolysis test and demonstrated no cytotoxicity, genotoxicity, and mutagenicity in the blood, marrow, and liver analyzes of the animals, corroborating data from the biochemical markers of GPT, GOT, and urea. Tissue regeneration was performed in all groups and was more pronounced in the group containing the combination of nHA/GNR (3%), which is consistent with the data obtained for the calcium, serum phosphorus, and ALP concentrations. Consequently, the study indicates that the engineered nanobiomaterial is a promising candidate for bone tissue repair and regenerative applications. STATEMENT OF SIGNIFICANCE: The scientific contribution of this study is the engineering of a synthetic hybrid biomaterial, in nanoscale by a pressing and heating process. A biodegradable polymeric matrix was covered on both sides with a carbonated hybrid bioceramic/graphene nanoribbons (GNR), which has hydrophilic characteristics, with chemical elements stoichiometrically similar to bone mineral composition. The nanomaterial displayed promising bone regeneration ability, which is the first example to be used in an osteoporotic animal model. Moreover, detailed biocompatibility and toxicity studies were performed on the nanomaterials and their compositions, which is of great interest for the scientific community.


Subject(s)
Durapatite , Nanotubes, Carbon , Animals , Biomineralization , Bone Regeneration , Rats , Tissue Engineering , Tissue Scaffolds
15.
J Funct Biomater ; 12(1)2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33562592

ABSTRACT

Electrospun ultrathin fibrous scaffold filed with synthetic nanohydroxyapatite (nHAp) and graphene nanoribbons (GNR) has bioactive and osteoconductive properties and is a plausible strategy to improve bone regeneration. Poly(butylene-adipate-co-terephthalate) (PBAT) has been studied as fibrous scaffolds due to its low crystallinity, faster biodegradability, and good mechanical properties; however, its potential for in vivo applications remains underexplored. We proposed the application of electrospun PBAT with high contents of incorporated nHAp and nHAp/GNR nanoparticles as bone grafts. Ultrathin PBAT, PBAT/nHAp, and PBAT/nHAp/GNR fibers were produced using an electrospinning apparatus. The produced fibers were characterized morphologically and structurally using scanning electron (SEM) and high-resolution transmission electron (TEM) microscopies, respectively. Mechanical properties were analyzed using a texturometer. All scaffolds were implanted into critical tibia defects in rats and analyzed after two weeks using radiography, microcomputed tomography, histological, histomorphometric, and biomechanical analyses. The results showed through SEM and high-resolution TEM characterized the average diameters of the fibers (ranged from 0.208 µm ± 0.035 to 0.388 µm ± 0.087) and nHAp (crystallite around 0.28, 0.34, and 0.69 nm) and nHAp/GNR (200-300 nm) nanoparticles distribution into PBAT matrices. Ultrathin fibers were obtained, and the incorporated nHAp and nHAp/GNR nanoparticles were well distributed into PBAT matrices. The addition of nHAp and nHAp/GNR nanoparticles improved the elastic modulus of the ultrathin fibers compared to neat PBAT. High loads of nHAp/GNR (PBATnH5G group) improved the in vivo lamellar bone formation promoting greater radiographic density, trabecular number and stiffness in the defect area 2 weeks after implantation than control and PBAT groups.

16.
Int J Nanomedicine ; 16: 667-682, 2021.
Article in English | MEDLINE | ID: mdl-33531806

ABSTRACT

BACKGROUND: Nanostructured surface modifications of Ti-based biomaterials are moving up from a highly-promising to a successfully-implemented approach to developing safe and reliable implants. METHODS: The study's main objective is to help consolidate the knowledge and identify the more suitable experimental strategies related to TiO2 nanotubes-modified surfaces. In this sense, it proposes the thorough investigation of two optimized nanotubes morphologies in terms of their biological activity (cell cytotoxicity, alkaline phosphatase activity, alizarin red mineralization test, and cellular adhesion) and their electrochemical behavior in simulated body fluid (SBF) electrolyte. Layers of small-short and large-long nanotubes were prepared and investigated in their amorphous and crystallized states and compared to non-anodized samples. RESULTS: Results show that much more than the surface area development associated with the nanotubes' growth; it is the heat treatment-induced change from amorphous to crystalline anatase-rutile structures that ensure enhanced biological activity coupled to high corrosion resistance. CONCLUSION: Compared to both non-anodized and amorphous nanotubes layers, the crystallized nano-structures' outstanding bioactivity was related to the remarkable increase in their hydrophilic behavior, while the enhanced electrochemical stability was ascribed to the thickening of the dense rutile barrier layer at the Ti surface beneath the nanotubes.


Subject(s)
Nanotubes/chemistry , Prostheses and Implants , Titanium/chemistry , Alkaline Phosphatase/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Survival , Corrosion , Electrochemistry , Electrolytes/chemistry , Humans , Nanotubes/ultrastructure , Wettability
17.
Commun Biol ; 4(1): 233, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33608611

ABSTRACT

The engineering of multifunctional surgical bactericidal nanofibers with inherent suitable mechanical and biological properties, through facile and cheap fabrication technology, is a great challenge. Moreover, hernia, which is when organ is pushed through an opening in the muscle or adjacent tissue due to damage of tissue structure or function, is a dire clinical challenge that currently needs surgery for recovery. Nevertheless, post-surgical hernia complications, like infection, fibrosis, tissue adhesions, scaffold rejection, inflammation, and recurrence still remain important clinical problems. Herein, through an integrated electrospinning, plasma treatment and direct surface modification strategy, multifunctional bactericidal nanofibers were engineered showing optimal properties for hernia repair. The nanofibers displayed good bactericidal activity, low inflammatory response, good biodegradation, as well as optimal collagen-, stress fiber- and blood vessel formation and associated tissue ingrowth in vivo. The disclosed engineering strategy serves as a prominent platform for the design of other multifunctional materials for various biomedical challenges.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biocompatible Materials , Gelatin/pharmacology , Hernia, Abdominal/surgery , Herniorrhaphy/instrumentation , Methacrylates/pharmacology , Nanofibers , Polyesters/pharmacology , Surgical Wound Infection/prevention & control , Tissue Scaffolds , Animals , Anti-Bacterial Agents/chemistry , Disease Models, Animal , Gelatin/chemistry , Hernia, Abdominal/pathology , Methacrylates/chemistry , Mice , NIH 3T3 Cells , Nanomedicine , Polyesters/chemistry , Rats , Surgical Wound Infection/microbiology , Wound Healing/drug effects
18.
Colloids Surf B Biointerfaces ; 194: 111192, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32599503

ABSTRACT

Cartilage is one of our body's tissues which are not repaired automatically by itself. Problems associated with cartilage are very common worldwide and are considered the leading cause of pain and disability. Smart biomaterial or "Four dimensional" (4D) biomaterials has started emerging as a suitable candidate, which are principally three dimensional (3D) materials that change their morphology or generate a response measured at space and time to physiologic stimuli. In this context, the release of oxygen through hydrogels in contact with water is considered as 4D biomaterials. The objective of this study is to develop strategies to release oxygen in a sustainable and prolonged manner through hydrogels systems to promote chondrocytes survival in oxygen-free environment. The 4D biomaterials are engineered from gelatin methacryloyl (GelMA) loaded with calcium peroxide (CPO), which have the ability to generate oxygen in a controlled and sustained manner for up to 6 days. The incorporation of CPO into the hydrogel system provided materials with enhanced mechanical and porosity properties. Furthermore, the hydrogels promoted chondrocyte survival and reduced cell death under oxygen-free conditions.


Subject(s)
Chondrocytes , Hydrogels , Gelatin , Oxygen , Tissue Engineering
19.
J Funct Biomater ; 11(1)2020 Feb 22.
Article in English | MEDLINE | ID: mdl-32098431

ABSTRACT

Stereolithography technology associated with the employment of photocrosslinkable, biocompatible, and bioactive hydrogels have been widely used. This method enables 3D microfabrication from images created by computer programs and allows researchers to design various complex models for tissue engineering applications. This study presents a simple and fast home-made stereolithography system developed to print layer-by-layer structures. Polyethylene glycol diacrylate (PEGDA) and gelatin methacryloyl (GelMA) hydrogels were employed as the photocrosslinkable polymers in various concentrations. Three-dimensional (3D) constructions were obtained by using the stereolithography technique assembled from a commercial projector, which emphasizes the low cost and efficiency of the technique. Lithium phenyl-2,4,6-trimethylbenzoyl phosphonate (LAP) was used as a photoinitiator, and a 404 nm laser source was used to promote the crosslinking. Three-dimensional and vascularized structures with more than 5 layers and resolutions between 42 and 83 µm were printed. The 3D printed complex structures highlight the potential of this low-cost stereolithography technique as a great tool in tissue engineering studies, as an alternative to bioprint miniaturized models, simulate vital and pathological functions, and even for analyzing the actions of drugs in the human body.

20.
Int J Biol Macromol ; 165(Pt B): 2813-2822, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33736284

ABSTRACT

Flowers-like ZnO structures were synthesized using Arabic Gum (AGZnO) or Karaya Gum (KGZnO). The AGZnO and KGZnO were characterized by X-ray diffractometry, Fourier Transformed Infrared, Scanning Electron Microscopy, Photoluminescence, nitrogen adsorption/desorption and diffuse reflectance techniques. The materials were tested in the discoloration of Methylene Blue (MB) dye under visible light and scavenger studies were also performed. The toxicity of the MB irradiated was investigated in bioassays with Artemia salina. The structural characterization demonstrated the formation of hexagonal ZnO. All samples presented flower-like morphology with presence of mesopores identified by BET method. The optical properties indicated band gap of 2.99 (AGZnO) and 2.76 eV (KGZnO), and emission in violet, blue and green emissions also were observed. The KGZnO demonstrated better photocatalytic performance than the AGZnO, and scavenger studies indicated that OH radicals are the main species involved in the degradation of the pollutant model. The photodiscoloration of MB solution did not demonstrate toxicity. Therefore, KGZnO is a promising material for photocatalysis application.


Subject(s)
Artemia/growth & development , Gum Arabic/chemistry , Karaya Gum/chemistry , Methylene Blue/analysis , Zinc Oxide/chemistry , Adsorption , Animals , Artemia/drug effects , Catalysis , Green Chemistry Technology , Light , Materials Testing , Microscopy, Electron, Scanning , Molecular Structure , Photolysis , X-Ray Diffraction , Zinc Oxide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...